Next Article in Journal
N-Doping/KOH Synergy in Waste Moss Biochar for Geosmin Removal in Aquaculture Water: Elucidating Surface Functionalization and Activation Mechanisms
Previous Article in Journal
Probiotic-Based Cleaning Solutions: From Research Hypothesis to Infection Control Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

C-Terminal Modification Contributes the Antibacterial Activity of a Cecropin-like Region of Heteroscorpine-1 from Scorpion Venom

by
Yutthakan Saengkun
1,2,
Anuwatchakij Klamrak
1,2,
Piyapon Janpan
1,2,
Shaikh Shahinur Rahman
1,3,
Rima Erviana
4,
Nawan Puangmalai
5,
Nisachon Jangpromma
2,6,
Jureerut Daduang
2,7,
Sakda Daduang
1,2 and
Jringjai Areemit
8,*
1
Division of Pharmacognosy and Toxicology, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
2
Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Khon Kaen University, Khon Kaen 40000, Thailand
3
Department of Applied Nutrition and Food Technology, Faculty of Biological Sciences, Islamic University, Kushtia 7000, Bangladesh
4
School of Pharmacy, Universitas Muhammadiyah Yogyakarta, Jl. Brawijaya, Tamantirto, Bantul, Yogyakarta 55183, Indonesia
5
Innovative Pharma Herbs Co., Ltd., 67/3 M.5, Tha Raeng, Ban Laem 76110, Thailand
6
Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen 40000, Thailand
7
Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
8
Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
*
Author to whom correspondence should be addressed.
Biology 2025, 14(8), 1044; https://doi.org/10.3390/biology14081044
Submission received: 23 June 2025 / Revised: 28 July 2025 / Accepted: 9 August 2025 / Published: 13 August 2025
(This article belongs to the Section Biochemistry and Molecular Biology)

Simple Summary

Antimicrobial peptides (AMPs) are promising candidates for combating multidrug-resistant bacteria; however, the production of large peptides is often expensive. In this study, the cecropin-like region of heteroscorpine-1 (CeHS-1) was modified using truncation, amino acid substitution, end-tagging, and amidation strategies. The results demonstrated that end-tagging with an arginine–tryptophan–tryptophan (RWW) stretch enhanced the antibacterial activity and DNA-binding properties of short CeHS-1 analogs without causing damage to human red blood cells. Newly designed CeHS-1 analogs strongly interacted with the grooves of DNA, as shown by molecular docking and in vitro assays. Our study clearly shows that the RWW end-tag plays a crucial role in determining the antibacterial activity of short AMPs, as confirmed by the wet lab results.

Abstract

The rise of multidrug-resistant pathogens has become a serious health concern, creating an urgent need for novel therapeutic approaches. Among the compounds explored, AMPs have emerged as promising candidates due to their broad-spectrum activity and low propensity for resistance development. However, their clinical implementation is limited by improper size, in vivo instability, and toxicity. Here, we designed short analogs of CeHS-1 via (1) truncation of intact CeHS-1, (2) amino acid substitution, (3) end-tagging, and (4) C-terminal amidation. The results showed that short analogs fused with an RWW stretch exhibited stronger antibacterial activity than the parent analogs, without inducing hemolysis in human red blood cells. Among the tested AMPs, mechanistic studies revealed membrane-disruptive activity of certain peptides against Staphylococcus aureus. In silico analyses also suggested that the analogs bind DNA by aligning parallel to its grooves, where the RWW stretch is believed to contribute to interactions between arginine and tryptophan residues and nitrogenous bases through electrostatic, hydrogen bonding, and hydrophobic interactions. The short CeHS-1 analogs established here may serve as potential alternative antimicrobial agents, which should be tested in clinical trials in the future.

1. Introduction

The global rise of multidrug-resistant (MDR) pathogens represents a critical public health emergency, with projections suggesting that antimicrobial resistance (AMR) could result in up to 10 million deaths annually by 2050 if left unmitigated [1,2,3]. This crisis is further exacerbated by a sharp decline in the development of new antibiotics [4]. As a result, alternative antimicrobial strategies are urgently needed. Antimicrobial peptides (AMPs), a group of naturally occurring defense molecules, have gained attention as promising candidates owing to their broad-spectrum efficacy, low cytotoxicity, and reduced risk of resistance development [5,6].
AMPs are typically composed of 10–50 amino acid residues and are produced by a wide array of organisms, including mammals, insects, amphibians, and plants [5,7,8,9]. These peptides play an integral role in innate immunity and display activity against bacteria, fungi, viruses, and protozoa [10,11]. Their antimicrobial function is largely attributed to their amphipathic structure and net positive charge ranging from +2 to +9, which enable selective interaction with negatively charged microbial membranes [12,13,14]. Hydrophobic domains promote membrane insertion and disruption through mechanisms such as pore formation and bilayer destabilization [15,16]. Moreover, AMPs may translocate across membranes and target intracellular components such as nucleic acids, further enhancing their bactericidal effect [5,17]. In addition to antimicrobial properties, certain AMPs have demonstrated anticancer potential, expanding their therapeutic applications [18].
Despite their therapeutic promise, AMPs face clinical translation challenges including proteolytic degradation, cytotoxicity, limited bioavailability, and high manufacturing costs [1,19]. To circumvent these limitations, rational design strategies such as sequence truncation, amino acid substitution, hydrophobic end-tagging, and C-terminal amidation have been employed to enhance peptide stability, potency, and specificity [20,21].
Short AMPs are particularly advantageous due to their ease of synthesis, structural simplicity, and favorable pharmacokinetics [22,23,24]. Notably, several truncated analogs of LL-37, such as P60.4Ac and SAAP-148, exhibit enhanced antibacterial activity and reduced toxicity, including efficacy against resistant pathogens [25,26]. Similarly, CeHS-1, derived from heteroscorpine-1 (HS-1) found in the venom of Heterometrus laoticus, has demonstrated potent antimicrobial activity, particularly against methicillin-resistant Staphylococcus aureus (MRSA) and multidrug-resistant Klebsiella pneumoniae [27].
Amino acid substitution is widely used to optimize AMP properties by modifying net charge, hydrophobicity, and amphipathicity [21,28,29]. Enhanced variants such as MEP-3 and MEP-4 outperform their parental peptide, melectin, in combating diverse bacterial strains [30]. Another optimization method, end-tagging with hydrophobic residues—especially stretches of tryptophan (Trp, W)—has been shown to improve peptide stability and antimicrobial potency [31,32,33]. Similarly, C-terminal amidation, a natural post-translational modification found in several animal-derived AMPs, neutralizes negative charges and boosts peptide–membrane interactions while protecting against carboxypeptidase-mediated degradation [34,35,36,37,38,39,40].
Venomous organisms such as scorpions represent a rich source of bioactive peptides, including AMPs with unique structural motifs and potent antimicrobial activity [41,42,43] CeHS-1, a 36-residue cecropin-like peptide located at the N-terminus of HS-1, possesses a typical cecropin structure composed of an amphipathic α-helical N-terminal region and a hydrophobic C-terminal segment. This peptide is active against both Gram-positive and Gram-negative bacteria, including resistant strains, while maintaining minimal hemolytic and cytotoxic activity [27]. However, large peptide size and associated production costs limit its broader application [44,45].
To address these challenges, the present study focused on the rational design of short CeHS-1 analogs by applying a systematic approach involving sequence truncation, targeted amino acid substitutions, end-tagging with an RWW tripeptide, and C-terminal amidation. The newly designed analogs were evaluated for their antibacterial efficacy, hemolytic activity, membrane disruption potential, and DNA-binding properties. The findings offer insight into the structure–activity relationships of short AMPs and highlight the therapeutic potential of cost-effective synthetic analogs derived from scorpion venom peptides.

2. Materials and Methods

2.1. Peptide Design

CeHS-1 was split at the hinge region (shown in Figure S1) into two fragments, F1-CeHS-1 and F2-CeHS-1. These fragments were then analyzed using the Antimicrobial Peptide Calculator and Predictor (available online at https://aps.unmc.edu/prediction) (accessed on 5 February 2023). The program revealed sequence homology between F1-CeHS-1 and F2-CeHS-1 and known potent AMPs. Each fragment was further divided at the midpoint, resulting in four shorter fragments: F1.1-CeHS-1, F1.2-CeHS-1, F2.1-CeHS-1, and F2.2-CeHS-1. These were subsequently compared to other short AMPs. Sequence alignment indicated a high degree of similarity between these smaller fragments and efficient short AMPs. Based on these results, the small fragments were used as templates for designing new CeHS-1 analogs.
The small fragments of CeHS-1 were analyzed for helical wheel projections and physicochemical properties using the HeliQuest service (http://heliquest.ipmc.cnrs.fr/cgi-bin/ComputParams.py) (accessed on 10 February 2023) to guide amino acid substitutions. Negatively charged residues (Asp and Glu) and some polar uncharged amino acids (Asn and Gln) were replaced with positively charged residues (Lys) and hydrophobic amino acids (Ile) to improve overall physicochemical properties and enhance amphipathic distribution in the helical wheel diagrams. Additionally, the hydrophobic and hydrophilic regions of the analogs were expanded by incorporating a tripeptide RWW stretch at the C-terminus. Furthermore, C-terminal amidation was performed to increase peptide stability against proteolytic degradation and to enhance antibacterial activity.

2.2. Peptide Synthesis

The modified CeHS-1 analogs were synthesized using Fmoc solid-phase peptide synthesis (SPPS) by GL Biochem (Shanghai) Ltd. (Shanghai, China) with a purity of ≥95%. The purity was further confirmed by reverse-phase high-performance liquid chromatography, and the molecular weight of the peptides was analyzed using mass spectrometry.

2.3. Antimicrobial Activity Assay

The following microorganisms were used to determine the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of the peptides: Gram-positive Staphylococcus aureus ATCC 25923, Gram-negative Escherichia coli ATCC 25922, Klebsiella pneumoniae ATCC 27736, and Pseudomonas aeruginosa ATCC 27853. The assays were conducted according to the protocol described by Erviana et al., with some modifications [27].
The antibacterial activity of the modified CeHS-1 analogs was evaluated using MIC and MBC assays. The MIC (minimum inhibitory concentration) is defined as the lowest concentration of antimicrobial peptides (AMPs) or other agents that inhibits visible bacterial growth. In contrast, the MBC (minimum bactericidal concentration) is defined as the lowest concentration of AMPs that kills 99.9% of the bacterial inoculum under the specified conditions [46]. The determination of MIC values was performed using a 96-well plate assay. Initially, peptides were serially diluted in the wells. Then, 10 µL of bacterial inoculum, previously adjusted to a cell density of 2.2 × 106 CFU/mL, was added to each well. The plates were incubated at 37 °C for 16–18 h. The MIC was determined by measuring the optical density at 600 nm (OD600), with the lowest concentration of AMPs that inhibited visible bacterial growth defined as the MIC. For MBC determination, 25 µL of culture from wells showing no visible growth (i.e., those at and above the MIC) was spread onto nutrient agar (NA) plates and incubated at 37 °C for 16–18 h. The lowest concentration of AMPs at which no bacterial colonies were observed was defined as the MBC [47].

2.4. Hemolytic Activity Assay

The hemolytic activity of the peptides was assessed using human red blood cells (hRBCs), following the method described by Erviana et al., with some modifications [27]. Briefly, a 4% hRBC suspension was prepared in 1X phosphate-buffered saline (PBS, pH 7.4). Subsequently, 100 µL of the hRBC suspension was mixed with 100 µL of peptide samples at various concentrations (500, 200, 100, 50, and 25 µg/mL) in microcentrifuge tubes. The mixture was incubated at 37 °C for 1 h, followed by centrifugation at 100× g for 5 min. The supernatant was collected and transferred to a 96-well plate for absorbance measurement at 415 nm. The hemolytic activity of each peptide was subsequently calculated using the following equation:
H e m o l y s i s   ( % ) = S P × 100
In the above equation, S represents the absorbance at 415 nm of the sample or negative control, while P represents the absorbance at 415 nm of the positive control. The peptide solvent was used as the negative control, whereas 0.1% Triton X-100 served as the positive control.

2.5. In Silico Prediction of Hemolytic Activity

The HAPPENN tool webserver (https://research.timmons.eu/happenn) (accessed on 10 April 2024) was used to predict the likelihood of AMP-induced hemolysis. The amino acid sequences of the effective peptides, including those with C-terminal amidation, were submitted to classify them as either hemolytic or non-hemolytic.

2.6. Membrane Disruption Property Assay

A bacterial suspension of S. aureus was cultured in LB medium to the exponential phase (OD600 = 0.5). The cell suspension was then diluted to 106 CFU/mL in PBS and incubated with AMPs at varying concentrations (0.25, 0.5, and 1 MIC) for 1 h at 37 °C with continuous shaking at 180 rpm. Following incubation, 42 µL of SYTOX™ Green Ready Flow™ Reagent (Invitrogen, Thermo Fisher Scientific, Carlsbad, CA, USA) was added to the peptide-treated cells and incubated for an additional 10 min at 25 °C. Fluorescence emission was measured using flow cytometry. The cell population was gated based on forward scatter and side scatter parameters. Cells with compromised membranes were identified by their fluorescence (excitation at 488 nm and detection at 530 nm with a 30 nm bandpass filter). The mean fluorescence intensity and the percentage of fluorescent cells were determined by analyzing 100,000 cells per sample. Each experiment was performed in at least triplicate. Flow cytometry measurements were conducted using a BD FACSCanto II system (BD Biosciences, Wokingham, UK).

2.7. DNA-Binding Assay

Gel retardation assays were employed to examine the binding of CeHS-1 and its analogs to DNA, based on the method described by Anunthawan et al., with minor modifications. Briefly, plasmid DNA (pET32a vector) was mixed with 10× binding buffer (100 mM Tris–HCl, pH 8.0; 200 mM KCl; 10 mM EDTA; 10 mM DTT; and 50% (v/v) glycerol) and peptides at increasing peptide-to-DNA weight ratios of 0:1, 0.25:1, 0.5:1, 1:1, and 2:1. The mixtures were incubated at 25 °C for 1 h, after which 6× DNA loading dye was added. Samples were then loaded onto a 0.75% agarose/0.5× Tris-acetate-EDTA (TAE) gel and electrophoresed at 75 V for 1 h.

2.8. Molecular Docking

Docking analysis was performed using the HADDOCK 2.4 webserver (https://rascar.science.uu.nl/haddock2.4/) (accessed on 20 November 2024) to explore the binding interactions between AMPs and a DNA fragment. The HADDOCK analysis generated binding scores for the peptide–DNA complexes, calculated as a weighted sum of various energy terms, including van der Waals energy, electrostatic energy, desolvation energy, and restraints violation energy. The 3D structures of all peptides were generated using AlphaFold2 (https://colab.research.google.com/github/sokrypton/ColabFold/blob/main/AlphaFold2.ipynb) (accessed on 14 August 2024). The DNA fragment (a 20-base-pair sequence) was prepared from the 3D structure of the phage 434 Cro/OR1 complex (PDB: 3CRO) [48], with the phage 434 Cro protein removed using PyMOL. All amino acid residues and nucleotides were defined as active residues, indicating direct involvement in the interaction. Docking parameters were set to their default values. PyMOL and Discovery Studio were utilized to visualize and illustrate the interactions between the antimicrobial peptides (AMPs) and the DNA fragment.

3. Results

3.1. Peptides Design and Physicochemical Properties

F1-CeHS-1 and F2-CeHS-1, the fragments separated at the flexible hinge region of CeHS-1, shared high similarities with several small peptides classified as alpha-helix AMPs, as shown in Table 1. The comparison revealed similarities in the positioning of certain hydrophobic, positively charged, and other residues, which are highlighted in yellow, blue, and grey, respectively.
In addition, the shorter analogs of F1-CeHS-1 (F1.1-CeHS-1, F1.2-CeHS-1) and F2-CeHS-1 (F2.1-CeHS-1 and F2.2-CeHS-1) also exhibited strong similarities to several short AMPs, as detailed in Table 2 and Table 3. The amino acid alignment revealed resemblances in the positioning of specific hydrophobic and positively charged residues. This suggests that these peptide analogs have the potential to function as AMPs.
To improve the short CeHS-1 analogs, the original peptides (F1.1-CeHS-1, F1.2-CeHS-1, F2.1-CeHS-1, and F2.2-CeHS-1) were modified through amino acid substitutions and C-terminal end-tagging with a RWW stretch. As shown in Table 4, these modifications enhanced the physicochemical properties of the peptides. Furthermore, addition of the RWW stretch at the C-terminus increased both the positive charge and hydrophobicity of the peptides. However, for peptide No. 3, this modification disrupted the amphipathic division, as illustrated in Figure 1.

3.2. Antimicrobial Activity

The modified CeHS-1 analogs (peptides No. 2, No. 3, No. 5, No. 6, No. 8, No. 9, No. 11, and No. 12) were evaluated for their antibacterial activities by determining the minimum inhibitory concentrations (MIC) and minimum bactericidal concentrations (MBC) against four bacterial strains. The MIC and MBC values in Table 5 reveal that the modified analogs with the added RWW stretch—specifically peptides No. 6, No. 9, and No. 12—exhibited inhibitory effects against all tested bacterial strains. In contrast, peptide No. 3 showed activity only against E. coli. The results also indicated that E. coli was most sensitive to peptide No. 12, while K. pneumoniae and P. aeruginosa showed the highest sensitivity to peptides No. 9 and No. 6, respectively. The parent peptides (peptide No. 2, No. 5, No. 8, and No. 11) showed no antibacterial activity against any of the tested strains.

3.3. Hemolytic Activity

Toxicity is one of the key factors limiting the application of AMPs. To assess the safety of these peptides, the hemolytic activity of modified CeHS-1 analogs with potent antimicrobial properties was evaluated using human red blood cells (hRBCs). As shown in Figure 2, peptide No. 3 exhibited dose-dependent hemolytic activity, whereas the other peptides displayed consistent hemolytic properties across the tested concentration range. At the highest concentration tested (500 µg/mL), peptide No. 3 showed the greatest hemolytic activity, with a value of 7.74 ± 1.18%. In contrast, peptides No. 6, No. 9, and No. 12 exhibited lower hemolytic activities, with values of 2.35 ± 0.20%, 2.39 ± 0.22%, and 1.67 ± 0.17%, respectively. These results are almost consistent with the hemolytic predictions from HAPPENN, as shown in Table 6, where peptide No. 3 had the highest PROB score (0.556), indicating a greater likelihood of causing hemolysis. Although, peptides No. 6 and No. 12 were predicted to have low PROB scores, they still exhibited similar average hemolytic activity to peptide No. 9, which had a PROB score of 0.262 based on the results, a score over 20 times greater than those of peptides No. 6 and No.12. This indicates that care must be taken when interpreting results. The computational analysis is useful for initial screening, while should be complemented by experimental validation to ensuring the accuracy of the findings.

3.4. Membrane Disruption Property

Many AMPs kill bacteria by inducing membrane disruption and causing leakage of cellular contents [69,70]. Therefore, we evaluated the ability of CeHS-1 analogs (No. 6, No. 9, and No. 12) to disrupt the cell membrane integrity of S. aureus—a well-known pathogen associated with a range of skin infections, including atopic dermatitis, impetigo, and wound infections [33]—using a fluorescence dye-based assay with the DNA-binding probe SYTOX™ Green. SYTOX™ Green is a high-affinity nucleic acid stain that becomes fluorescent only upon binding to nucleic acids and cannot penetrate intact cell membranes. This property enables the distinction between non-permeabilized and permeabilized cells by detecting increased fluorescence when the dye binds to DNA [70,71,72]. Ampicillin and kanamycin were used as negative control compounds, as these antimicrobial agents exert their effects without disrupting the bacterial cell membrane [73,74].
The results presented in Figure 3 reveal that peptides No. 6 and No. 9 disrupt the cytoplasmic membrane of S. aureus in a dose-dependent manner, as indicated by increased fluorescence intensity. In contrast, peptide No. 12 exhibited consistently low fluorescence intensity, similar to that observed with the negative control compounds, ampicillin and kanamycin.

3.5. DNA-Binding Property

As previously mentioned, AMPs can penetrate bacterial cell membranes and bind to intracellular targets, particularly DNA, thereby disrupting DNA replication and RNA synthesis, which ultimately leads to cell death [5,17]. Thereby DNA could be another target of inhibition. To prove this hypothesis, the DNA-binding abilities of CeHS-1 and its analogs were evaluated using plasmid DNA (pET32a vector) as the target. The results presented in Figure 4 indicate that peptides incorporating the RWW stretch, specifically peptides No. 3, No. 6, No. 9, and No. 12, effectively inhibited DNA migration at a 1:1 plasmid DNA-to-peptide weight ratio. In contrast, CeHS-1 and the other peptides did not interfere with DNA migration, even at the highest weight ratio tested. These findings highlight that incorporating the RWW stretch significantly enhances the DNA-binding properties of the peptides. Furthermore, the results demonstrate that these peptides can bind to multiple forms of plasmid DNA.

3.6. Molecular Docking for DNA-Binding Property

Molecular docking studies were performed using the HADDOCK 2.4 webserver to elucidate the interactions between DNA and the CeHS-1 analogs. As shown in Table 7, CeHS-1 analogs lacking the RWW stretch (No. 2, No. 5, No. 8, and No. 11) exhibited binding scores ranging from −37.6 to −54.4 kcal/mol, which are higher than those of Magainin 2, a peptide known for its weak DNA-binding properties. In contrast, CeHS-1 analogs containing the RWW stretch (No. 3, No. 6, No. 9, and No. 12) demonstrated lower binding scores, ranging from −69.3 to −80.0 kcal/mol. These values are lower than those of Magainin 2 and are comparable to RT2 and KT2, antimicrobial peptides known to bind bacterial DNA to exert their effects. The results suggest that incorporating the RWW stretch at the C-terminus significantly enhances the DNA-binding ability of the peptides, while analogs lacking this modification exhibit weaker binding. This finding is consistent with the gel retardation assay results.
In addition, molecular docking analysis (Figure 5) revealed binding interactions between the peptide and DNA, with the peptide aligning parallel to the grooves of the DNA structure. Further interaction analysis (Table 7) showed that positively charged amino acids (Lys, Arg, and His) mainly interacted with the phosphate groups through hydrogen bonding and electrostatic interactions. Conversely, hydrophobic aromatic amino acids such as Trp and Phe primarily interacted with the nitrogenous bases via hydrogen bonding and hydrophobic interactions. These findings suggest that adding the RWW stretch at the C-terminus enhances the DNA-binding ability of CeHS-1 by increasing interactions with both the nitrogenous bases and the phosphate backbone of the DNA structure.

4. Discussion

Although CeHS-1 demonstrates potent antimicrobial properties, its full-length structure poses challenges in pharmaceutical development due to high production costs associated with synthetic manufacturing. While recombinant expression in Escherichia coli has been explored as an alternative production method, this strategy has yielded limited success due to reduced antimicrobial activity and low peptide yield [75]. In response to these challenges, this study employed rational peptide engineering to design shorter analogs of CeHS-1 that retain or exceed the activity of the parent peptide while minimizing cytotoxic effects and reducing synthesis costs.
Generally, the amino acid sequences of AMPs can be divided into two regions—hydrophobic and hydrophilic—that are typically of similar size. Many studies have attempted to improve the antibacterial activity of peptides by tagging them with hydrophobic oligopeptides, such as Trp and phenylalanine (Phe, F) [32,33]. Building on this strategy, Anunthawan et al. developed a systematic approach to enhance antibacterial activity of Luecrocin I isolated from the white blood cells of Crocodylus siamensis by simultaneously introducing both hydrophobic and hydrophilic regions through the incorporation of specific amino acid stretches and found exceptional enhanced antimicrobial activities of the modified peptide. In their design (ABBABBAABB/B), “A” denotes positively charged amino acids (e.g., Lys), while “B” denotes hydrophobic amino acids (e.g., Typ). However, both CRT2 and CRT3 demonstrated approximately threefold-higher hemolytic toxicity compared with the original Luecrocin I, likely due to the increased hydrophobicity resulting from the incorporation of these long stretches [31]. This bring us to use the RWW stretch—the shortest repeating amino acid motif—as it is more suitable for our objective of developing a systematic approach to designing short peptides and minimizing the risk of hemolytic activity associated with high hydrophobicity.
The systematic approach employed—comprising sequence truncation, amino acid substitution, C-terminal RWW end-tagging, and amidation—resulted in several analogs with enhanced antibacterial activity and physicochemical properties. The addition of the RWW tripeptide stretch significantly improved both the net positive charge and hydrophobicity of the analogs (as shown in Table 4), thereby promoting bacterial membrane interaction and disruption. These findings align with previous research demonstrating that the spatial arrangement of hydrophobic and cationic residues is critical for AMP efficacy [31,32]. However, the structural integrity and amphipathic division of the peptide remain essential, as evidenced by peptide No. 3, which exhibited lower antimicrobial activity despite RWW end-tagging—likely due to an unfavorable amphipathic profile.
Among the analogs, peptides No. 6, No. 9, and No. 12 emerged as the most promising candidates, demonstrating broad-spectrum antimicrobial activity at low MIC values (4–32 µg/mL) without inducing hemolysis in human red blood cells. These analogs thus satisfy key pharmacological criteria, including selectivity toward microbial membranes and minimal toxicity to host cells. Computational predictions using the HAPPENN tool further validated their low hemolytic potential, offering a cost- and time-efficient strategy for future AMP screening.
Mechanistic assays provided insight into the dual mode of action exhibited by these analogs. Peptides No. 6 and No. 9 disrupted bacterial membranes in a dose-dependent manner, as evidenced by increased SYTOX Green fluorescence, indicative of cytoplasmic membrane permeabilization. Interestingly, peptide No. 12 did not exhibit significant membrane-disruptive effects but instead displayed strong DNA-binding activity, suggesting an intracellular target-based mechanism. These findings are consistent with previous studies indicating that AMPs can act via both membrane-lytic and intracellular modes, including interference with DNA replication and transcription [5,17,76].
Gel retardation and molecular docking analyses confirmed the DNA-binding capabilities of the RWW-tagged peptides. Docking scores for peptides No. 6, No. 9, and No. 12 were comparable to known DNA-targeting AMPs such as RT2 and KT2. The RWW motif appeared to facilitate multiple electrostatic and hydrophobic interactions with phosphate backbones and nitrogenous bases, thus stabilizing peptide–DNA binding. These interactions highlight the structural and functional advantages of RWW incorporation, providing a framework for future AMP design.
Despite these promising results, large-scale production remains a hurdle. While chemical synthesis is feasible for research and clinical testing, it is not cost-effective for mass production. As such, developing a microbial expression system for the production of these short CeHS-1 analogs is a logical next step. This approach could offer advantages in scalability, speed, and cost, especially if combined with downstream purification strategies optimized for short, cationic peptides [77,78].
In summary, this study underscores the potential of rationally designed short AMPs as viable alternatives to traditional antibiotics, particularly in the context of rising drug resistance. By combining structural bioinformatics, in vitro assays, and molecular docking, this study presents a robust model for peptide optimization that can be extended to other AMP scaffolds.

5. Conclusions

In this study, we successfully designed small CeHS-1 analogs through a combination of sequence shortening, amino acid substitution, end-tagging with an appended RWW stretch, and C-terminal amidation. As demonstrated, the short CeHS-1 analogs containing the RWW stretch (peptides No. 3, No. 6, No. 9, and No. 12) exhibited promising antibacterial activity across various pathogenic bacteria while inducing no hemolysis in hRBCs. These analogs demonstrated DNA-binding ability, as suggested by molecular docking, and possibly displayed membrane-disruptive activity against S. aureus. Furthermore, our study suggests that the addition of the RWW stretch significantly enhances the antibacterial activity and DNA-binding capacity of CeHS-1 analogs by improving their physicochemical properties via enhanced positive charge and hydrophobicity.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/biology14081044/s1. Figure S1: 3D structure of CeHS-1. Table S1: The amino acid sequences of the AMPs mentioned in the Introduction and Discussion sections [25,26,27,29,30].

Author Contributions

Conceptualization, Y.S., A.K., S.D. and J.A.; methodology, N.J., Y.S., A.K. and R.E.; software, Y.S. and P.J.; validation, Y.S. and P.J.; formal analysis, Y.S. and A.K.; investigation, Y.S. and A.K.; resources, R.E., N.P., N.J., S.D. and J.A.; data curation, Y.S.; writing—original draft preparation, Y.S., A.K., S.S.R. and J.A.; writing—review and editing, Y.S., A.K., S.S.R. and J.A.; visualization, Y.S. and P.J.; supervision, J.D., S.D. and J.A.; project administration, J.D., S.D. and J.A.; funding acquisition, N.P., S.D. and J.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand (Contract No. 2-(2)/2563). This research was also funded by the National Research Council of Thailand (NRCT). This research was additionally funded by The Fundamental Fund of Khon Kean University (KKU), which received financial support from the National Science, Research and Innovation Fund (NSRF), Thailand. The research was supported by NSRF under the Basic Research Fund of Khon Kaen University. This research was funded by Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by Khon Kaen University Ethics Committee for Human Research—protocol code: HE642059, approval date: 21 July 2021.

Informed Consent Statement

Informed consent has been obtained from the patient to publish this paper.

Data Availability Statement

All data supporting the conclusions of this article are included in this article.

Conflicts of Interest

Author Nawan Puangmalai is a co-founder of Innovative Pharma Herbs Co., Ltd. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Mwangi, J.; Kamau, P.M.; Thuku, R.C.; Lai, R. Design Methods for Antimicrobial Peptides with Improved Performance. Zool. Res. 2023, 44, 1095–1114. [Google Scholar] [CrossRef]
  2. Antimicrobial Resistance Collaborators. Global Burden of Bacterial Antimicrobial Resistance in 2019: A Systematic Analysis. Lancet 2022, 399, 629–655. [Google Scholar] [CrossRef]
  3. de Kraker, M.E.A.; Stewardson, A.J.; Harbarth, S. Will 10 Million People Die a Year Due to Antimicrobial Resistance by 2050? PLoS Med. 2016, 13, e1002184. [Google Scholar] [CrossRef] [PubMed]
  4. Ventola, C.L. The Antibiotic Resistance Crisis: Part 1: Causes and Threats. Pharm. Ther. 2015, 40, 277–283. [Google Scholar]
  5. Dong, C.; Li, M.; Zhang, R.; Lu, W.; Xu, L.; Liu, J.; Chu, X. The Expression of Antibacterial Peptide Turgencin A in Pichia Pastoris and an Analysis of Its Antibacterial Activity. Molecules 2023, 28, 5405. [Google Scholar] [CrossRef] [PubMed]
  6. Chen, N.; Jiang, C. Antimicrobial Peptides: Structure, Mechanism, and Modification. Eur. J. Med. Chem. 2023, 255, 115377. [Google Scholar] [CrossRef]
  7. da Cunha, N.B.; Cobacho, N.B.; Viana, J.F.C.; Lima, L.A.; Sampaio, K.B.O.; Dohms, S.S.M.; Ferreira, A.C.R.; de la Fuente-Núñez, C.; Costa, F.F.; Franco, O.L.; et al. The next Generation of Antimicrobial Peptides (AMPs) as Molecular Therapeutic Tools for the Treatment of Diseases with Social and Economic Impacts. Drug Discov. Today 2017, 22, 234–248. [Google Scholar] [CrossRef]
  8. Watkins, R.R.; Bonomo, R.A. Overview: Global and Local Impact of Antibiotic Resistance. Infect. Dis. Clin. N. Am. 2016, 30, 313–322. [Google Scholar] [CrossRef]
  9. Ma, X.; Wang, Q.; Ren, K.; Xu, T.; Zhang, Z.; Xu, M.; Rao, Z.; Zhang, X. A Review of Antimicrobial Peptides: Structure, Mechanism of Action, and Molecular Optimization Strategies. Fermentation 2024, 10, 540. [Google Scholar] [CrossRef]
  10. Asensio-Calavia, P.; González-Acosta, S.; Otazo-Pérez, A.; López, M.R.; Morales-delaNuez, A.; Pérez de la Lastra, J.M. Teleost Piscidins-in Silico Perspective of Natural Peptide Antibiotics from Marine Sources. Antibiotics 2023, 12, 855. [Google Scholar] [CrossRef]
  11. Sinha, R.; Shukla, P. Antimicrobial Peptides: Recent Insights on Biotechnological Interventions and Future Perspectives. Protein Pept. Lett. 2019, 26, 79–87. [Google Scholar] [CrossRef]
  12. Huerta-Cantillo, J.; Navarro-García, F. Properties and Design of Antimicrobial Peptides as Potential Tools against Pathogens and Malignant Cells. Investig. Discapac. 2016, 5, 96–115. [Google Scholar]
  13. Rungsa, P.; Peigneur, S.; Jangpromma, N.; Klaynongsruang, S.; Tytgat, J.; Daduang, S. In Silico and in Vitro Structure-Activity Relationship of Mastoparan and Its Analogs. Molecules 2022, 27, 561. [Google Scholar] [CrossRef] [PubMed]
  14. Tajer, L.; Paillart, J.C.; Dib, H.; Sabatier, J.M.; Fajloun, Z.; Abi Khattar, Z. Molecular Mechanisms of Bacterial Resistance to Antimicrobial Peptides in the Modern Era: An Updated Review. Microorganisms 2024, 12, 1259. [Google Scholar] [CrossRef] [PubMed]
  15. Lee, T.-H.; Hall, K.N.; Aguilar, M.-I. Antimicrobial Peptide Structure and Mechanism of Action: A Focus on the Role of Membrane Structure. Curr. Top. Med. Chem. 2016, 16, 25–39. [Google Scholar] [CrossRef] [PubMed]
  16. Gagat, P.; Ostrówka, M.; Duda-Madej, A.; Mackiewicz, P. Enhancing Antimicrobial Peptide Activity through Modifications of Charge, Hydrophobicity, and Structure. Int. J. Mol. Sci. 2024, 25, 10821. [Google Scholar] [CrossRef]
  17. Hilpert, K.; McLeod, B.; Yu, J.; Elliott, M.R.; Rautenbach, M.; Ruden, S.; Bürck, J.; Muhle-Goll, C.; Ulrich, A.S.; Keller, S.; et al. Short Cationic Antimicrobial Peptides Interact with ATP. Antimicrob. Agents Chemother. 2010, 54, 4480–4483. [Google Scholar] [CrossRef]
  18. Gaspar, D.; Veiga, A.S.; Castanho, M.A.R.B. From Antimicrobial to Anticancer Peptides. a Review. Front. Microbiol. 2013, 4, 294. [Google Scholar] [CrossRef] [PubMed]
  19. Kim, H.; Jang, J.H.; Kim, S.C.; Cho, J.H. De Novo Generation of Short Antimicrobial Peptides with Enhanced Stability and Cell Specificity. J. Antimicrob. Chemother. 2014, 69, 121–132. [Google Scholar] [CrossRef]
  20. Li, J.; Koh, J.-J.; Liu, S.; Lakshminarayanan, R.; Verma, C.S.; Beuerman, R.W. Membrane Active Antimicrobial Peptides: Translating Mechanistic Insights to Design. Front. Neurosci. 2017, 11, 73. [Google Scholar] [CrossRef]
  21. Huan, Y.; Kong, Q.; Mou, H.; Yi, H. Antimicrobial Peptides: Classification, Design, Application and Research Progress in Multiple Fields. Front. Microbiol. 2020, 11, 582779. [Google Scholar] [CrossRef]
  22. Mikut, R.; Ruden, S.; Reischl, M.; Breitling, F.; Volkmer, R.; Hilpert, K. Improving Short Antimicrobial Peptides despite Elusive Rules for Activity. Biochim. Biophys. Acta 2016, 1858, 1024–1033. [Google Scholar] [CrossRef] [PubMed]
  23. Mangoni, M.L.; Shai, Y. Short Native Antimicrobial Peptides and Engineered Ultrashort Lipopeptides: Similarities and Differences in Cell Specificities and Modes of Action. Cell. Mol. Life Sci. 2011, 68, 2267–2280. [Google Scholar] [CrossRef] [PubMed]
  24. Sharma, L.; Bisht, G.S. Short Antimicrobial Peptides: Therapeutic Potential and Recent Advancements. Curr. Pharm. Des. 2023, 29, 3005–3017. [Google Scholar] [CrossRef] [PubMed]
  25. Nell, M.J.; Tjabringa, G.S.; Wafelman, A.R.; Verrijk, R.; Hiemstra, P.S.; Drijfhout, J.W.; Grote, J.J. Development of Novel LL-37 Derived Antimicrobial Peptides with LPS and LTA Neutralizing and Antimicrobial Activities for Therapeutic Application. Peptides 2006, 27, 649–660. [Google Scholar] [CrossRef]
  26. de Breij, A.; Riool, M.; Cordfunke, R.A.; Malanovic, N.; de Boer, L.; Koning, R.I.; Ravensbergen, E.; Franken, M.; van der Heijde, T.; Boekema, B.K.; et al. The Antimicrobial Peptide SAAP-148 Combats Drug-Resistant Bacteria and Biofilms. Sci. Transl. Med. 2018, 10, eaan4044. [Google Scholar] [CrossRef]
  27. Erviana, R.; Saengkun, Y.; Rungsa, P.; Jangpromma, N.; Tippayawat, P.; Klaynongsruang, S.; Daduang, J.; Daduang, S. Novel Antimicrobial Peptides from a Cecropin-like Region of Heteroscorpine-1 from Heterometrus Laoticus Venom with Membrane Disruption Activity. Molecules 2021, 26, 5872. [Google Scholar] [CrossRef]
  28. Torres, M.D.T.; Sothiselvam, S.; Lu, T.K.; de la Fuente-Nunez, C. Peptide Design Principles for Antimicrobial Applications. J. Mol. Biol. 2019, 431, 3547–3567. [Google Scholar] [CrossRef]
  29. Lee, J.-K.; Park, S.-C.; Hahm, K.-S.; Park, Y. Antimicrobial HPA3NT3 Peptide Analogs: Placement of Aromatic Rings and Positive Charges Are Key Determinants for Cell Selectivity and Mechanism of Action. Biochim. Biophys. Acta 2013, 1828, 443–454. [Google Scholar] [CrossRef]
  30. Cerovský, V.; Hovorka, O.; Cvacka, J.; Voburka, Z.; Bednárová, L.; Borovicková, L.; Slaninová, J.; Fucík, V. Melectin: A Novel Antimicrobial Peptide from the Venom of the Cleptoparasitic Bee Melecta Albifrons. Chembiochem 2008, 9, 2815–2821. [Google Scholar] [CrossRef]
  31. Anunthawan, T.; Yaraksa, N.; Phosri, S.; Theansungnoen, T.; Daduang, S.; Dhiravisit, A.; Thammasirirak, S. Improving the Antibacterial Activity and Selectivity of an Ultra Short Peptide by Hydrophobic and Hydrophilic Amino Acid Stretches. Bioorg. Med. Chem. Lett. 2013, 23, 4657–4662. [Google Scholar] [CrossRef]
  32. Schmidtchen, A.; Pasupuleti, M.; Mörgelin, M.; Davoudi, M.; Alenfall, J.; Chalupka, A.; Malmsten, M. Boosting Antimicrobial Peptides by Hydrophobic Oligopeptide End Tags. J. Biol. Chem. 2009, 284, 17584–17594. [Google Scholar] [CrossRef]
  33. Pasupuleti, M.; Schmidtchen, A.; Chalupka, A.; Ringstad, L.; Malmsten, M. End-Tagging of Ultra-Short Antimicrobial Peptides by W/F Stretches to Facilitate Bacterial Killing. PLoS ONE 2009, 4, e5285. [Google Scholar] [CrossRef]
  34. Gan, B.H.; Gaynord, J.; Rowe, S.M.; Deingruber, T.; Spring, D.R. The Multifaceted Nature of Antimicrobial Peptides: Current Synthetic Chemistry Approaches and Future Directions. Chem. Soc. Rev. 2021, 50, 7820–7880. [Google Scholar] [CrossRef]
  35. de Santana, C.J.C.; Pires Júnior, O.R.; Fontes, W.; Palma, M.S.; Castro, M.S. Mastoparans: A Group of Multifunctional α-Helical Peptides with Promising Therapeutic Properties. Front. Mol. Biosci. 2022, 9, 824989. [Google Scholar] [CrossRef]
  36. Mangoni, M.L.; Fiocco, D.; Barra, D.; Simmaco, M. Bombinins. In Handbook of Biologically Active Peptides; Academic Press: Cambridge, MA, USA, 2006; pp. 333–337. [Google Scholar] [CrossRef]
  37. Shahmiri, M.; Mechler, A. The Role of C-Terminal Amidation in the Mechanism of Action of the Antimicrobial Peptide Aurein 1.2. Eurobiotech J. 2020, 4, 25–31. [Google Scholar] [CrossRef]
  38. Van Wyk, R.J.; Serem, J.C.; Oosthuizen, C.B.; Semenya, D.; Serian, M.; Lorenz, C.D.; Mason, A.J.; Bester, M.J.; Gaspar, A.R.M. Carboxy-Amidated AamAP1-Lys Has Superior Conformational Flexibility and Accelerated Killing of Gram-Negative Bacteria. Biochemistry 2025, 64, 841–859. [Google Scholar] [CrossRef]
  39. Nikookar Golestani, R.; Ghods, E.; Rostamian, M.; Madanchi, H.; Talebi, A.F. Investigating the Antimicrobial Activity, Cytotoxicity, and Action Mechanism of Acylated and Amidated Derivatives of AurH1 Antifungal Peptide. BMC Microbiol. 2023, 23, 332. [Google Scholar] [CrossRef] [PubMed]
  40. Salazar-Hernández, D.; Juárez-González, V.R.; Bustamante, V.H.; Martínez-Martínez, L.L.; Ramírez, V.; Balleza, D.; Quintero-Hernández, V. Conformational Flexibility and Net Charge Are Key Determinants for the Antimicrobial Activity of Peptide Uy234 Against Multidrug-Resistant Bacteria. Int. J. Pept. Res. Ther. 2024, 30, 79. [Google Scholar] [CrossRef]
  41. Roy, A.; Bharadvaja, N. Venom-Derived Bioactive Compounds as Potential Anticancer Agents: A Review. Int. J. Pept. Res. Ther. 2021, 27, 129–147. [Google Scholar] [CrossRef]
  42. Kawakami, H.; Goto, S.G.; Murata, K.; Matsuda, H.; Shigeri, Y.; Imura, T.; Inagaki, H.; Shinada, T. Isolation of Biologically Active Peptides from the Venom of Japanese Carpenter Bee, Xylocopa Appendiculata. J. Venom. Anim. Toxins Incl. Trop. Dis. 2017, 23, 29. [Google Scholar] [CrossRef] [PubMed]
  43. Bordon, K.D.C.F.; Cologna, C.T.; Fornari-Baldo, E.C.; Pinheiro-Júnior, E.L.; Cerni, F.A.; Amorim, F.G.; Anjolette, F.A.P.; Cordeiro, F.A.; Wiezel, G.A.; Cardoso, I.A.; et al. From Animal Poisons and Venoms to Medicines: Achievements, Challenges and Perspectives in Drug Discovery. Front. Pharmacol. 2020, 11, 1132. [Google Scholar] [CrossRef]
  44. Garvey, M. Antimicrobial Peptides Demonstrate Activity against Resistant Bacterial Pathogens. Infect. Dis. Rep. 2023, 15, 454–469. [Google Scholar] [CrossRef]
  45. Dijksteel, G.S.; Ulrich, M.M.W.; Middelkoop, E.; Boekema, B.K.H.L. Review: Lessons Learned from Clinical Trials Using Antimicrobial Peptides (AMPs). Front. Microbiol. 2021, 12, 616979. [Google Scholar] [CrossRef] [PubMed]
  46. Balouiri, M.; Sadiki, M.; Ibnsouda, S.K. Methods for in Vitro Evaluating Antimicrobial Activity: A Review. J. Pharm. Anal. 2016, 6, 71–79. [Google Scholar] [CrossRef]
  47. Al-Ani, I.; Zimmermann, S.; Reichling, J.; Wink, M. Pharmacological Synergism of Bee Venom and Melittin with Antibiotics and Plant Secondary Metabolites against Multi-Drug Resistant Microbial Pathogens. Phytomedicine 2015, 22, 245–255. [Google Scholar] [CrossRef]
  48. Mondragón, A.; Harrison, S.C. The Phage 434 CroOR1 Complex at 2.5 Å Resolution. J. Mol. Biol. 1991, 219, 321–334. [Google Scholar] [CrossRef]
  49. Rozek, T.; Wegener, K.L.; Bowie, J.H.; Olver, I.N.; Carver, J.A.; Wallace, J.C.; Tyler, M.J. The Antibiotic and Anticancer Active Aurein Peptides from the Australian Bell Frogs Litoria Aurea and Litoria Raniformis the Solution Structure of Aurein 1.2. Eur. J. Biochem. 2000, 267, 5330–5341. [Google Scholar] [CrossRef]
  50. Qi, X.; Zhou, C.; Li, P.; Xu, W.; Cao, Y.; Ling, H.; Ning Chen, W.; Ming Li, C.; Xu, R.; Lamrani, M.; et al. Novel Short Antibacterial and Antifungal Peptides with Low Cytotoxicity: Efficacy and Action Mechanisms. Biochem. Biophys. Res. Commun. 2010, 398, 594–600. [Google Scholar] [CrossRef] [PubMed]
  51. Dos Santos Cabrera, M.P.; De Souza, B.M.; Fontana, R.; Konno, K.; Palma, M.S.; De Azevedo, W.F.; Ruggiero Neto, J. Conformation and Lytic Activity of Eumenine Mastoparan: A New Antimicrobial Peptide from Wasp Venom. J. Pept. Res. 2004, 64, 95–103. [Google Scholar] [CrossRef] [PubMed]
  52. Prates, M.V.; Sforça, M.L.; Regis, W.C.B.; Leite, J.R.S.A.; Silva, L.P.; Pertinhez, T.A.; Araújo, A.L.T.; Azevedo, R.B.; Spisni, A.; Bloch, C. The NMR-Derived Solution Structure of a New Cationic Antimicrobial Peptide from the Skin Secretion of the Anuran Hyla Punctata. J. Biol. Chem. 2004, 279, 13018–13026. [Google Scholar] [CrossRef]
  53. Wu, S.; Nie, Y.; Zeng, X.C.; Cao, H.; Zhang, L.; Zhou, L.; Yang, Y.; Luo, X.; Liu, Y. Genomic and Functional Characterization of Three New Venom Peptides from the Scorpion Heterometrus Spinifer. Peptides 2014, 53, 30–41. [Google Scholar] [CrossRef]
  54. Liu, R.; Liu, H.; Ma, Y.; Wu, J.; Yang, H.; Ye, H.; Lai, R. There Are Abundant Antimicrobial Peptides in Brains of Two Kinds of Bombina Toads. J. Proteome Res. 2011, 10, 1806–1815. [Google Scholar] [CrossRef]
  55. Csordás, A.; Michl, H. Isolierung Und Strukturaufklärung Eines Hämolytisch Wirkenden Polypeptides Aus Dem Abwehrsekret Europäischer Unken. Monatsh Chem. 1970, 101, 182–189. [Google Scholar] [CrossRef]
  56. Yang, X.; Lee, W.-H.; Zhang, Y. Extremely Abundant Antimicrobial Peptides Existed in the Skins of Nine Kinds of Chinese Odorous Frogs. J. Proteome Res. 2012, 11, 306–319. [Google Scholar] [CrossRef]
  57. King, J.D.; Al-Ghaferi, N.; Abraham, B.; Sonnevend, A.; Leprince, J.; Nielsen, P.F.; Conlon, J.M. Pentadactylin: An Antimicrobial Peptide from the Skin Secretions of the South American Bullfrog Leptodactylus Pentadactylus. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2005, 141, 393–397. [Google Scholar] [CrossRef]
  58. Thompson, A.H.; Bjourson, A.J.; Orr, D.F.; Shaw, C.; McClean, S. A Combined Mass Spectrometric and CDNA Sequencing Approach to the Isolation and Characterization of Novel Antimicrobial Peptides from the Skin Secretions of Phyllomedusa Hypochondrialis Azurea. Peptides 2007, 28, 1331–1343. [Google Scholar] [CrossRef]
  59. Bradford, A.M.; Raftery, M.J.; Bowie, J.H.; Tyler, M.J.; Wallace, J.C.; Adams, G.W.; Severini, C. Novel Uperin Peptides from the Dorsal Glands of the Australian Flood-Plain Toadlet Uperoleia Inundata. Aust. J. Chem. Int. J. Chem. Sci. 1996, 49, 475–484. [Google Scholar] [CrossRef]
  60. Ouyang, X.; Li, B.; Yang, T.; Yang, Y.; Ba, Z.; Zhang, J.; Yang, P.; Liu, Y.; Wang, Y.; Zhao, Y.; et al. High Therapeutic Index α-Helical AMPs and Their Therapeutic Potential on Bacterial Lung and Skin Wound Infections. ACS Infect. Dis. 2024, 10, 3138–3157. [Google Scholar] [CrossRef] [PubMed]
  61. Wang, G.; Watson, K.M.; Peterkofsky, A.; Buckheit, R.W. Identification of Novel Human Immunodeficiency Virus Type 1-Inhibitory Peptides Based on the Antimicrobial Peptide Database. Antimicrob. Agents Chemother. 2010, 54, 1343–1346. [Google Scholar] [CrossRef] [PubMed]
  62. Monincová, L.; Buděšínský, M.; Slaninová, J.; Hovorka, O.; Cvačka, J.; Voburka, Z.; Fučík, V.; Borovičková, L.; Bednárová, L.; Straka, J.; et al. Novel Antimicrobial Peptides from the Venom of the Eusocial Bee Halictus Sexcinctus (Hymenoptera: Halictidae) and Their Analogs. Amino Acids 2010, 39, 763–775. [Google Scholar] [CrossRef]
  63. Hong, W.; Li, T.; Song, Y.; Zhang, R.; Zeng, Z.; Han, S.; Zhang, X.; Wu, Y.; Li, W.; Cao, Z. Inhibitory Activity and Mechanism of Two Scorpion Venom Peptides against Herpes Simplex Virus Type 1. Antivir. Res. 2014, 102, 1–10. [Google Scholar] [CrossRef]
  64. Yang, X.; Hu, Y.; Xu, S.; Hu, Y.; Meng, H.; Guo, C.; Liu, Y.; Liu, J.; Yu, Z.; Wang, H. Identification of Multiple Antimicrobial Peptides from the Skin of Fine-Spined Frog, Hylarana Spinulosa (Ranidae). Biochimie 2013, 95, 2429–2436. [Google Scholar] [CrossRef] [PubMed]
  65. Richter, A.; Sutherland, D.; Ebrahimikondori, H.; Babcock, A.; Louie, N.; Li, C.; Coombe, L.; Lin, D.; Warren, R.L.; Yanai, A.; et al. Associating Biological Activity and Predicted Structure of Antimicrobial Peptides from Amphibians and Insects. Antibiotics 2022, 11, 1710. [Google Scholar] [CrossRef]
  66. Lee, J.; Kang, H.K.; Cheong, H.; Park, Y. A Novel Antimicrobial Peptides From Pine Needles of Pinus Densiflora Sieb. et Zucc. Against Foodborne Bacteria. Front. Microbiol. 2021, 12, 662462. [Google Scholar] [CrossRef]
  67. Gause, G.F.; Brazhnikova, M.G. Gramicidin S and Its Use in the Treatment of Infected Wounds. Nature 1944, 154, 703. [Google Scholar] [CrossRef]
  68. Cantisani, M.; Leone, M.; Mignogna, E.; Kampanaraki, K.; Falanga, A.; Morelli, G.; Galdiero, M.; Galdiero, S. Structure-Activity Relations of Myxinidin, an Antibacterial Peptide Derived from the Epidermal Mucus of Hagfish. Antimicrob. Agents Chemother. 2013, 57, 5665–5673. [Google Scholar] [CrossRef] [PubMed]
  69. Sandhu, G.; Morrow, M.R.; Booth, V. Roles of Histidine Charge and Cardiolipin in Membrane Disruption by Antimicrobial Peptides Gaduscidin-1 and Gaduscidin-2. Biochim. Biophys. Acta (BBA)-Biomembr. 2020, 1862, 183444. [Google Scholar] [CrossRef] [PubMed]
  70. Benfield, A.H.; Henriques, S.T. Mode-of-Action of Antimicrobial Peptides: Membrane Disruption vs. Intracellular Mechanisms. Front. Med. Technol. 2020, 2, 610997. [Google Scholar] [CrossRef]
  71. Rathinakumar, R.; Walkenhorst, W.F.; Wimley, W.C. Broad-Spectrum Antimicrobial Peptides by Rational Combinatorial Design and High-Throughput Screening: The Importance of Interfacial Activity. J. Am. Chem. Soc. 2009, 131, 7609–7617. [Google Scholar] [CrossRef]
  72. Sani, M.A.; Henriques, S.T.; Weber, D.; Separovic, F. Bacteria May Cope Differently from Similar Membrane Damage Caused by the Australian Tree Frog Antimicrobial Peptide Maculatin 1.1. J. Biol. Chem. 2015, 290, 19853–19862. [Google Scholar] [CrossRef] [PubMed]
  73. Kaushik, D.; Mohan, M.; Borade, D.M.; Swami, O.C. Ampicillin: Rise Fall and Resurgence. J. Clin. Diagn. Res. 2014, 8, ME01. [Google Scholar] [CrossRef] [PubMed]
  74. Mingeot-Leclercq, M.P.; Glupczynski, Y.; Tulkens, P.M. Aminoglycosides: Activity and Resistance. Antimicrob. Agents Chemother. 1999, 43, 727–737. [Google Scholar] [CrossRef]
  75. Erviana, R.; Saengkun, Y.; Rungsa, P.; Jangpromma, N.; Mustofa; Daduang, S. The Recombinant Expression and Antimicrobial Activity Determination of Cecropin-like Part of Heteroscorpine-1 from Heterometrus Laoticus. Biodiversitas 2022, 23, 5646–5653. [Google Scholar] [CrossRef]
  76. Le, C.-F.; Fang, C.-M.; Sekaran, S.D. Intracellular Targeting Mechanisms by Antimicrobial Peptides. Antimicrob. Agents Chemother. 2017, 61, 10-1128. [Google Scholar] [CrossRef]
  77. Zhang, J.; Movahedi, A.; Wei, Z.; Sang, M.; Wu, X.; Wang, M.; Wei, H.; Pan, H.; Yin, T.; Zhuge, Q. High-Level SUMO-Mediated Fusion Expression of ABP-DHC-Cecropin A from Multiple Joined Genes in Escherichia coli. Anal. Biochem. 2016, 509, 15–23. [Google Scholar] [CrossRef]
  78. Chubukov, V.; Mukhopadhyay, A.; Petzold, C.J.; Keasling, J.D.; Martín, H.G. Synthetic and Systems Biology for Microbial Production of Commodity Chemicals. NPJ Syst. Biol. Appl. 2016, 2, 16009. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Helical wheel projections of CeHS-1 analogs.
Figure 1. Helical wheel projections of CeHS-1 analogs.
Biology 14 01044 g001
Figure 2. Hemolytic activity of CeHS-1 analogs against human red blood cells after 1 h exposure.
Figure 2. Hemolytic activity of CeHS-1 analogs against human red blood cells after 1 h exposure.
Biology 14 01044 g002
Figure 3. Cytoplasmic membrane permeabilization of S. aureus evaluated from SYTOX™ Green fluorescence emission intensity.
Figure 3. Cytoplasmic membrane permeabilization of S. aureus evaluated from SYTOX™ Green fluorescence emission intensity.
Biology 14 01044 g003
Figure 4. Peptide binding to plasmid DNA was assessed using a 0.75% agarose/TAE gel, with varying peptide-to-plasmid weight ratios. The ratios tested were 0:1, 0.25:1, 0.5:1, 1:1, and 2:1, displayed from left to right. The first lane, labeled ‘0’, represents the negative control with no peptide added. R represents the position of the relaxed circular plasmid, L denotes the linear plasmid, and S indicates the supercoiled plasmid.
Figure 4. Peptide binding to plasmid DNA was assessed using a 0.75% agarose/TAE gel, with varying peptide-to-plasmid weight ratios. The ratios tested were 0:1, 0.25:1, 0.5:1, 1:1, and 2:1, displayed from left to right. The first lane, labeled ‘0’, represents the negative control with no peptide added. R represents the position of the relaxed circular plasmid, L denotes the linear plasmid, and S indicates the supercoiled plasmid.
Biology 14 01044 g004
Figure 5. Illustrates a hypothetical interaction between CeHS-1 analogs and DNA fragment.
Figure 5. Illustrates a hypothetical interaction between CeHS-1 analogs and DNA fragment.
Biology 14 01044 g005
Table 1. Sequence alignment of CeHS-1 and its analogs with AMPs.
Table 1. Sequence alignment of CeHS-1 and its analogs with AMPs.
PeptideOriginSequenceAAIdentity (%)APD IDRef.
CeHS-1 GWINEEK-IQKKIDEK-IGNNILG---G---MA-K-AV-VHK-LAKG--E-F-Q37
F1-CeHS-1This workGWINEEK-IQKKIDEK-IG
Aurein 1.1Litoria aureaG---LFD-IIKKIAES-I- 1341.18AP00012[49]
P11-5Synthetic peptideG-----K-LFKKIL-K-IL 1141.18AP03683[50]
Mastoparan-AFAnterhynchium flavomarginatum micado--INLLK-IAKGII-K-SL 1441.18AP01517[51]
P11-6Synthetic peptide-----KKLI-KKIL-K-IL 1138.89AP03684[50]
Hylaseptin P1Hyla punctataG-I-LDA-IA-KIA-KAAG 1438.89AP01249[52]
SpiniferinHeterometrus spinifer--ILGE--IWKGI--KDIL 1338.89AP02551[53]
F2-CeHS-1This work NNILG---G---MA-K-AV-VHK-LAKG--E-F-Q21
Maximin 31Bombina maxima -GI-G---GALLSAGKSA---LKGLAKGLAEHF--2543.33AP01732[54]
BombininBombina variegata L -GI-----GAL-SA-KGA---LKGLAKGLAEHFAN2442.86AP00049[55]
Nigrocin-1-OW3Odorrana wuchuanensis -GILGNIVG---M-GKK-V-VCG-L-SGLC 2141.67AP01883[56]
Ocellatin-P1Leptodactylus pentadactylus -GLLDTLKG---AA-KN-V-V-GSLASKVME--L-2540.00AP00540[57]
Phylloseptin-H11Phyllomedusa hypochondrialis FLSL--IP----HAI-NAVGVH---AK----HF--1939.13AP00972[58]
Uperin 3.6Uperoleia inundata GVI-AA-KK-V-VNV-L-KNL---F 1738.10AP00325[59]
The yellow, blue, and grey backgrounds indicate the conserved hydrophobic, positively charged, and other amino acid residues, respectively.
Table 2. Sequence alignment of F1-CeHS-1 and its analogs with AMPs.
Table 2. Sequence alignment of F1-CeHS-1 and its analogs with AMPs.
PeptideOriginSequenceAAIdentity (%)APD IDRef.
F1-CeHS-1 G-W-INEEK-IQKKIDE--K-IG17
F1.1-CeHS-1This workG-W-INEEK-IQKK 11
IK2Synthetic peptideG---I-IKK-IIKKI 1050.00AP04801[60]
B6Synthetic peptideGIWSDLAE--IIKKF 1342.86AP03851[61]
Halictine 2Halictus sexcinctusGKW-MSLLK-ILK 1238.46AP01923[62]
F1.2-CeHS-1This work -K-IQKKIDE--K-IG11
IK2Synthetic peptide -G-IIKKIIK--K-I-1054.55AP04801[60]
P11-6Synthetic peptide KKLI-KKI-L--K-IL1153.85AP03684[50]
Hp1036Heterometrus petersii ---ILGKIWEGIKSIF1342.86AP02334[63]
The yellow, blue, and grey backgrounds indicate the conserved hydrophobic, positively charged, and other amino acid residues, respectively.
Table 3. Sequence alignment of F2-CeHS-1 and its analogs with AMPs.
Table 3. Sequence alignment of F2-CeHS-1 and its analogs with AMPs.
PeptideOriginSequenceAAIdentity (%)APD IDRef.
F2-CeHS-1 NNI--L-G--G-MA--KAVVHK-L--AK-GEFQ-21
F2.1-CeHS-1This workNNI--L G--G-MA--KAV 11
Temporin-SN2Hylarana spinulosa-FITGLIG--GLM---KAL 1346.67AP02273[64]
Temporin-SN3Hylarana spinulosa-FISGLIG--GLM---KAL 1346.67AP02274[64]
OdMa4Odorrana margaretae-GI--LSGLLG--AGKKIVC 1541.18AP03539[65]
F2.2-CeHS-1This work VHK-L--AK-GEFQ-10
PN5Pinus densiflora -FKFL--ARTGKFL 1141.67AP03449[66]
Gramicidin SBacillus brevis V-K-LFPVK--LFQ-1041.69AP02243[67]
MD4KMyxine glutinosa L. GIHKIL---KYGKPS-1238.46AP04009[68]
The yellow, blue, and grey backgrounds indicate the conserved hydrophobic, positively charged, and other amino acid residues, respectively.
Table 4. Amino acid sequences and physicochemical properties of amidated CeHS-1 analogs.
Table 4. Amino acid sequences and physicochemical properties of amidated CeHS-1 analogs.
No.Peptide NameAmino Acid SequenceAANet
Charge *
%HHµH
1F1.1-CeHS-1GWINEEKIQKK-NH211+222.270.0710.215
2F1.1-CeHS-1 IIGWINIIKIQKK-NH211+445.450.5150.497
3F1.1-CeHS-1 II-RTGWINIIKIQKKRWW-NH214+550.000.6540.461
4F1.2-CeHS-1KIQKKIDEKIG-NH211+322.27−0.0170.609
5F1.2-CeHS-1 IKKIQKKIIKKIG-NH211+636.360.1850.691
6F1.2-CeHS-1 IK-RTKIQKKIIKKIGRWW-NH214+742.860.3940.814
7F2.1-CeHS-1NNILGGMAKAV-NH211+254.550.3980.575
8F2.1-CeHS-1 KKKKILGGMAKAV-NH211+455.550.3270.618
9F2.1-CeHS-1 KK-RTKKILGGMAKAVRWW-NH214+557.140.5060.622
10F2.2-CeHS-1VHKLAKGEFQ-NH210+240.000.2310.394
11F2.2-CeHS-1 IKVHKLAKGIFK-NH210+450.000.3980.697
12F2.2-CeHS-1 IK-RTVHKLAKGIFKRWW-NH213+553.850.5750.654
Green, red, and blue represent the original amino acids, substituted amino acids, and tripeptide stretches, respectively. %H: hydrophobic amino acid content, H: hydrophobicity, µH: hydrophobic moment. * Amidation at the C-terminus increases the net charge of a peptide by one positive unit (+1).
Table 5. Determination of MIC of CeHS-1 analogs.
Table 5. Determination of MIC of CeHS-1 analogs.
No.Peptide NameS. aureusE. coliK. pneumoniaeP. aeruginosa
MICMBCMICMBCMICMBCMICMBC
2F1.1-CeHS-1 IINDNDNDNDNDNDNDND
3F1.1-CeHS-1 II-RTNDND128256NDNDNDND
5F1.2-CeHS-1 IKNDNDNDNDNDNDNDND
6F1.2-CeHS-1 IK-RT3264882562561632
8F2.1-CeHS-1 KKNDNDNDNDNDNDNDND
9F2.1-CeHS-1 KK-RT6412883212812864128
11F2.2-CeHS-1 IKNDNDNDNDNDNDNDND
12F2.2-CeHS-1 IK-RT32324162562563264
Positive control
Ampicillin88432>256>256128256
MIC: minimum inhibitory concentration. MBC: minimum bactericidal concentration. MIC and MBC values are determined in µg/Ml. ND indicates no detection at the highest tested concentration.
Table 6. Potential hemolytic activity of CeHS-1 analogs identified using HAPPENN tool.
Table 6. Potential hemolytic activity of CeHS-1 analogs identified using HAPPENN tool.
No.Peptide NamePROB
3F1.1-CeHS-1 II-RT0.556
6F1.2-CeHS-1 IK-RT0.007
9F2.1-CeHS-1 KK-RT0.262
12F2.2-CeHS-1 IK-RT0.010
The PROB score is a normalized sigmoid score ranging from 0 to 1. A score of 0 is predicted to be most likely non-hemolytic, and a score of 1 is predicted to be most likely hemolytic.
Table 7. Binding scores (in kcal/mol) and molecular interaction of CeHS-1 analogs and DNA fragment.
Table 7. Binding scores (in kcal/mol) and molecular interaction of CeHS-1 analogs and DNA fragment.
NoPeptide NameBinding Energy
(Kcal/mol)
RMSDChemical Bond Interaction (Å)/Nucleotide Composition
H-Bonds
Conventional H-Bond
Carbon H-Bond
Pi-Donor H-bond
Charge
Salt Bridge; Charge-Charge
Charge-Charge
π-Cation
π-Anion
Hydrophobic Interactions
Alkyl
π-alkyl
π-π shaped
2F1.1 CeHS-1 II−37.6 ± 4.5−5.2 ± 1.0Trp2P/P, Ile3D, Lys7P/P, Lys10G/T/T/D, Lys11PTrp2P, Lys7P/P, Lys11PN.D
3F1.1 CeHS-1 II-RT−71.8 ± 3.5−9.0 ± 0.8Lys7P, Gln9P, Lys10D/A/C/TTrp2●●P, Lys7P/P, Lys11P/P,Trp14T
5F1.2-CeHS-1 IK−47.3 ± 10.9−7.4 ± 0.6Lys5●●T, Lys9A/C/A/D, Ile10DLys1P/P, Lys4PN.D
6F1.2-CeHS-1 IK-RT−80.0 ± 6.3−8.7 ± 0.1Lys5T/D, Lys9C/T, Arg12P, Trp13P, Trp14PLys1P, Lys4P, Lys8P/P, Arg12●●P/P, N.D
8F2.1-CeHS-1 KK−41.7 ± 12.7−2.3 ± 1.4 Lys1P, Lys2C/P/D, Gly6D, Lys9T/C/C/●●DLys1P/PN.D
9F2.1-CeHS-1 KK-RT−79.4 ± 7.3−5.9 ± 0.3Lys1P, Lys2T/A, Lys9ALys1P/●●P, Lys2C/P, Lys9P/P, Arg12P/P, Trp14PLys2●●A/A, Lys9A
11F2.2-CeHS-1 IK−42.8 ± 3.7−5.0 ± 0.9His2T, Lys6T/G/D, Lys10T/D/PVal1P, Lys3●●P/P, Phe9PN.D
12F2.2-CeHS-1 IK-RT−69.3 ± 2.9−4.7 ± 0.9His2P, Lys6P, Lys10P, Trp13AVal1P, His2P, Lys6P/P, Lys10P/P, Trp12●●PVal1A, Ala5●●A, Phe9A/A, Trp13●●A/A
Positive control
RT2−81.7 ± 15.01.0 ± 0.9
KT2−90.5 ± 14.62.1 ± 1.2
Negative control
Magainin 2−59.4 ± 9.910.6 ± 0.5
N.D.: not detected, P: phosphate group, D: deoxyribose sugar, nitrogenous base; A: Adenine, G: Guanine, C: Cytosine, and T: Thymine.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Saengkun, Y.; Klamrak, A.; Janpan, P.; Rahman, S.S.; Erviana, R.; Puangmalai, N.; Jangpromma, N.; Daduang, J.; Daduang, S.; Areemit, J. C-Terminal Modification Contributes the Antibacterial Activity of a Cecropin-like Region of Heteroscorpine-1 from Scorpion Venom. Biology 2025, 14, 1044. https://doi.org/10.3390/biology14081044

AMA Style

Saengkun Y, Klamrak A, Janpan P, Rahman SS, Erviana R, Puangmalai N, Jangpromma N, Daduang J, Daduang S, Areemit J. C-Terminal Modification Contributes the Antibacterial Activity of a Cecropin-like Region of Heteroscorpine-1 from Scorpion Venom. Biology. 2025; 14(8):1044. https://doi.org/10.3390/biology14081044

Chicago/Turabian Style

Saengkun, Yutthakan, Anuwatchakij Klamrak, Piyapon Janpan, Shaikh Shahinur Rahman, Rima Erviana, Nawan Puangmalai, Nisachon Jangpromma, Jureerut Daduang, Sakda Daduang, and Jringjai Areemit. 2025. "C-Terminal Modification Contributes the Antibacterial Activity of a Cecropin-like Region of Heteroscorpine-1 from Scorpion Venom" Biology 14, no. 8: 1044. https://doi.org/10.3390/biology14081044

APA Style

Saengkun, Y., Klamrak, A., Janpan, P., Rahman, S. S., Erviana, R., Puangmalai, N., Jangpromma, N., Daduang, J., Daduang, S., & Areemit, J. (2025). C-Terminal Modification Contributes the Antibacterial Activity of a Cecropin-like Region of Heteroscorpine-1 from Scorpion Venom. Biology, 14(8), 1044. https://doi.org/10.3390/biology14081044

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop