Back to Journals » Infection and Drug Resistance » Volume 17

The Efficacy of Antimicrobial Peptides Against Gentamicin-Resistant Klebsiella pneumoniae [Letter]

Authors Mardiana M 

Received 26 June 2024

Accepted for publication 5 July 2024

Published 8 July 2024 Volume 2024:17 Pages 2815—2816

DOI https://doi.org/10.2147/IDR.S484430

Checked for plagiarism Yes

Editor who approved publication: Professor Sandip Patil



Meity Mardiana

Eijkman Research Center for Molecular Biology, National Research and Innovation Agency (BRIN), Cibinong, West Java, Indonesia

Correspondence: Meity Mardiana, Email [email protected]


View the original paper by Dr Chen and colleagues


Dear editor

Klebsiella pneumoniae is a Gram-negative, non-motile, and encapsulated bacteria found in the environment. Once this bacterium enters the body, it can exhibit a high degree of virulence.1 The frequent misuse of antibiotics has led to an increase in antibiotic-resistant cases of K. pneumoniae, complicating treatment efforts. Consequently, there is an urgent need for alternative therapies. In the study presented by Chen et al, the authors effectively utilized antimicrobial peptides (AMPs) to treat gentamicin-resistant K. pneumoniae. Currently, AMPs are extensively explored due to their ubiquitous presence in nature, their relatively short residue length, and their well-documented activity in killing both Gram-positive and Gram-negative bacteria through interactions with outer membrane structures and lipids, as well as intracellular structures such as nucleic acids and proteins, thereby disrupting bacterial cell functions.2 Chen et al revealed that in the gentamicin-resistant K. pneumoniae, genes related to efflux pump, porin, and biofilm formation, as well as the minimum inhibitory concentration (MIC), significantly increased. Furthermore, the study also provided evidence that AMPs can mitigate these genetic changes.3 Although this study demonstrated promising results, we believe there are aspects that can be improved. The authors used ATCC13883 as wild-type (WT) strain in this research. While it does not exhibit any antibiotic resistance profile, a comparison with the gradient-un-induced strain would yield more accurate data as there might be other future differences between the ATCC strain and the un-induced strain that could potentially influence the experiment outcomes. The nomenclature for the strains used in this study should also be revised in the data presentation to enhance reader comprehension. For example, use C for the gentamicin-induced strain and C’ for the gentamicin-induced strain. Furthermore, the authors conducted MIC assays on planktonic bacteria but did not perform a minimum biofilm inhibitory concentration (MBIC) assay on their biofilm experiments. While the percentage of biofilm inhibition assessed in this study is indeed important for evaluating the efficacy of AMPs in inhibiting biofilm formation, MBIC is equally crucial, as it is essential for determining the lowest concentration of AMPs used for therapy and for establishing precise dosages for future applications.4 Additionally, the authors could also investigate the ability of AMPs to eradicate biofilm formation, as in clinical settings, many cases involve mature biofilms that are more resistant to antibiotic treatment. Therefore, it is important to ascertain whether AMPs can effectively disrupt mature biofilms as well.

We appreciate the authors for their comprehensive study on the use of AMPs in combating antibiotic-resistant bacterial infections. Yet this area still faces significant limitations, including limited in vivo safety data and the potential for bacterial resistant development.5 Additionally, effective AMPs must exhibit characteristics such as low-cost production, environmental stability, and low serum binding capacity.6 These factors can also be explored by the authors in the future so that they can provide effective AMPs for clinical applications.

Disclosure

The author reports no conflicts of interest in this communication.

References

1. Ashurst JV, Dawson A. Klebsiella Pneumonia. Treasure Island (FL): StatPearls; 2024.

2. de Souza CM, da Silva AP, Junior NGO, Martinez OF, Franco OL. Peptides as a therapeutic strategy against Klebsiella pneumoniae. Trends Pharmacol Sci. 2022;43(4):335–348. doi:10.1016/j.tips.2021.12.006

3. Chen X, Zhang B, He J, et al. Exploration of antimicrobial peptides in the treatment of gentamicin-resistant Klebsiella pneumoniae Infection. Infect Drug Resist. 2024;17:2591–2605. doi:10.2147/IDR.S462653

4. Macia MD, Rojo-Molinero E, Oliver A. Antimicrobial susceptibility testing in biofilm-growing bacteria. Clin Microbiol Infect. 2014;20(10):981–990. doi:10.1111/1469-0691.12651

5. Kidd TJ, Mills G, Sa-Pessoa J, et al. A Klebsiella pneumoniae antibiotic resistance mechanism that subdues host defences and promotes virulence. EMBO Mol Med. 2017;9(4):430–447. doi:10.15252/emmm.201607336

6. Li J, Koh JJ, Liu S, Lakshminarayanan R, Verma CS, Beuerman RW. Membrane active antimicrobial peptides: translating mechanistic insights to design. Front Neurosci. 2017;11:73. doi:10.3389/fnins.2017.00073

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.