Skip to main content

EDITORIAL article

Front. Microbiol., 06 June 2023
Sec. Infectious Agents and Disease
This article is part of the Research Topic Burkholderia spp.-Transmission, Pathogenesis, Host-pathogen interaction, Prevention and Treatment View all 6 articles

Editorial: Burkholderia spp.-transmission, pathogenesis, host-pathogen interaction, prevention and treatment

  • 1Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
  • 2Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore

Bacteria belonging to the Burkholderia genus are rod-shaped, Gram-negatives, non-spore-forming, obligate aerobics. They can be found in the environment and are able to cause infections in plants and in animals, where they affect especially immunocompromised patients (Rodríguez-Cisneros et al., 2023). Within this Research Topic, different aspects related to Burkholderia spp. were investigated and discussed.

One of the main problems is related to the treatments that can be used to cure the infections caused by these bacteria. Indeed, most strains are multidrug resistant (MDR) (Mahenthiralingam et al., 2008), and this poses a serious threat in the management of infections. MDR phenotypes have been mainly ascribed to class A β-lactamases (Rhodes and Schweizer, 2016), penicillinase with broad spectrum activity (Hwang and Kim, 2015), efflux pump over-expression (Perrin et al., 2010; Bazzini et al., 2011), mutations in the drug target and reduced permeability (Scoffone et al., 2017).

Pattinson et al. used a multi-omic approach to investigate the mechanism of action of 12- cationic bola-amphiphile, 12,12′-(dodecane-1,12-diyl) bis (9-amino-1,2,3,4-tetrahydroacridinium), named 12-bis-THA against Burkholderia thailandensis. They compared the efficacy of the compound against capsulated and unencapsulated strains and no differences were observed in the minimum inhibitory concentration values. On the other hand, time-kill analysis revealed that the unencapsulated strain was more susceptible. So the authors decided to use proteomic and metabolomic tools to better understand how the compound works. Their findings suggest that 12-bis-THA affects the central metabolism and suppresses the production of the F1 domain of ATP synthase.

An alternative to fight drug resistance is represented by bioactive compounds, which are basically secondary metabolites (Elshafie et al., 2017). Natural products have higher chemical novelty than that synthetic drugs and are proving a boon in pharmaceutical industries together with molecular informatics, which has come up as an important tool in finding the relationships between molecules and their biological effects (Bender and Glen, 2004). The study by Prasad et al. is based on the screening of soil pathogens exhibiting strong antimicrobial activity, identifying bioactive compounds, elucidating the mode of action of compounds in an antimicrobial assay using in silico techniques, and identifying the likely targets through molecular docking studies. A total of 53 bacterial isolates were screened for their antimicrobial potential: the strains JRBHU6, which showed the highest antimicrobial property, was identified as Burkholderia seminalis. The strain produced hydrolytic enzymes (chitinases/cellulose) of significance in accrediting its antimicrobial effects. The bioactive metabolites produced by the isolate were extracted in different organic solvents and exerted the most potent growth inhibitory effects against multi-drug resistant Staphylococcus aureus and against fungal strains such as Fusarium oxysporum, Aspergillus niger, Microsporum gypseum, Trichophyton mentagrophytes, Trichoderma harzianum. This bioactive metabolite was identified as pyrrolo (1,2-a)pyrazine-1, 4-dione, hexahydro, pyrrolo(1,2-a)pyrazine-1,4-dione, hexahydro-3(2-methylpropyl).

Another important point to find alternative solutions to treat Burkholderia infections is to fully understand the immune response triggered by the presence of bacteria.

The study by Zhang et al. investigated the adaptive immune responses of dairy cows suffering from mastitis caused by Burkholderia contaminans. They mainly used flow cytometry to evaluate T- and B-lymphocytes-mediated immune responses in cows infected by B. contaminans and compared it with healthy animals. A significant a significant increase was observed in IgG+CD27+ B lymphocytes, in γδ T cells and double positive CD4+CD8+ T cells and the relative cytokines, and in activated WC1+ γδ T cells in the B. contaminans infected cows, indicating a major role of these cells against B. contaminans infection. On the contrary, the decreased number of B lymphocytes in infected animals suggests that humoral immune response may not be adequate to fight intracellular infections. All together these results shed light on the immune response triggered by the presence of Burkholderia bacteria in animals: this is important especially for the development of a vaccine, which is still lacking, that could help to prevent these infections thus avoiding the overuse of antibiotics in cattle that strongly contributes to the insurgence of MDR strains.

Other two articles of this Research Topic focused on Burkholderia pseudomallei, the causative agent of melioidosis. Melioidosis is an important seasonal infectious disease in tropical as well as sub-tropical counties (Suputtamongkol et al., 1994; Liu et al., 2015).

In the first study, the regulation of virulence factors involved in surface attachment was studied (Sun et al.). Different techniques were used, including high-throughput transposon mutagenesis screening, RNA-Seq analysis, qRT-PCR, and the results revealed that the protein SapR is a transcriptional regulator that activates the surface attachment protein Sap1, as well as genes associated with the bacterial membrane in response to diverse environments, including those encoding virulence factors important for the intracellular survival of B. pseudomallei.

In several parts of the world, including South West India, most of the clinical cases of human infection are reported during the time of heavy rainfall. However, the underlying root causes of this phenomenon are not clearly understood yet. India is one of the counties with the highest predicted cases of melioidosis burden worldwide. Surprisingly, there is very little information on the environmental distribution of B. pseudomallei as well as its main determining factors.

The study by Shaw et al. clearly reported that the prevalence of B. pseudomallei found in soil, especially in South West India, was determined by geochemical factors. They collected a total of 1,704 soil samplings from 20 diverse agricultural sites, particularly during the monsoon as well as post-monsoon season/summer, and screened them by quantitative real-time PCR (qPCR). Results of direct qPCR detected B. pseudomallei in all 20 sites. Further, B. pseudomallei DNA-positive samples were negatively linked with the concentration of iron, and manganese/nitrogen. The study clearly proves that South West India showed a wide environmental distribution of B. pseudomallei but also considerable differences in the abundance between sites and within single sites. In addition, nutrient-depleted habitats promote the presence of B. pseudomallei. Most importantly, the study confirmed that the very high levels of B. pseudomallei abundance in the soil are found during the rainy season. Indeed, most of the cases were clinically diagnosed during the monsoon season (Vidyalakshmi et al., 2012; Mukhopadhyay et al., 2018).

Author contributions

Both authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

Conflict of interest

The 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.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Bazzini, S., Udine, C., Sass, A., Pasca, M. R., Longo, F., Emiliani, G., et al. (2011). Deciphering the role of RND efflux transporters in Burkholderia cenocepacia. PLoS ONE. 6, e18902. doi: 10.1371/journal.pone.0018902

PubMed Abstract | CrossRef Full Text | Google Scholar

Bender, A., and Glen, R. C. (2004). Molecular similarity: a key technique in molecular informatics. Org. Biomol. Chem. 2, 3204–3218. doi: 10.1039/b409813g

PubMed Abstract | CrossRef Full Text | Google Scholar

Elshafie, H. S., Viggiani, L., Mostafa, M. S., El-Hashash, M. A., Bufo, S. A., and Camele, I. (2017). Biological activity and chemical identification of ornithine lipid produced by Burkholderia gladioli pv. Agaricicola ICMP 11096 using LC-MS and NMR analyses. J. Biol. Res. 90, 96–103. doi: 10.4081/jbr.2017.6534

CrossRef Full Text | Google Scholar

Hwang, J., and Kim, H. S. (2015). Cell wall recycling-linked coregulation of AmpC and PenB β-Lactamases through ampD mutations in Burkholderia cenocepacia. Antimicrob. Agents Chemother. 59, 7602–7610. doi: 10.1128/aac.01068-15

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, X., Pang, L., Sim, S. H., Goh, K. T., Ravikumar, S., Win, M. S., et al. (2015). Association of melioidosis incidence with rainfall and humidity, Singapore, 2003-2012. Emerg. Infect. Dis. 21, 159–162. doi: 10.3201/eid2101.140042

PubMed Abstract | CrossRef Full Text | Google Scholar

Mahenthiralingam, E., Baldwin, A., and Dowson, C. G. (2008). Burkholderia cepacia complex bacteria: opportunistic pathogens with important natural biology. J. Appl. Microbiol. 104, 1539–1551. doi: 10.1111/j.1365-2672.2007.03706.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Mukhopadhyay, C., Shaw, T., Varghese, G. M., and Dance, D. A. B. (2018). Melioidosis in South Asia (India, Nepal, Pakistan, Bhutan and Afghanistan). Trop. Med. Infect. Dis. 3, 51. doi: 10.3390/tropicalmed3020051

PubMed Abstract | CrossRef Full Text | Google Scholar

Perrin, E., Fondi, M., Papaleo, M. C., Maida, I., Buroni, S., Pasca, M. R., et al. (2010). Exploring the HME and HAE1 efflux systems in the genus Burkholderia. BMC Evol Biol. 10, 164. doi: 10.1186/1471-2148-10-164

PubMed Abstract | CrossRef Full Text | Google Scholar

Rhodes, K. A., and Schweizer, H. P. (2016). Antibiotic resistance in Burkholderia species. Drug Resist. Updat. 28, 82–90. doi: 10.1016/j.drup.2016.07.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodríguez-Cisneros, M., Morales-Ruíz, L. M., Salazar-Gómez, A., Rojas-Rojas, F. U., and Estrada-de Los Santos, P. (2023). Compilation of the antimicrobial compounds produced by Burkholderia Sensu Stricto. Molecules. 28, 1646. doi: 10.3390/molecules28041646

PubMed Abstract | CrossRef Full Text | Google Scholar

Scoffone, V. C., Chiarelli, L. R., Trespidi, G., Mentasti, M., Riccardi, G., and Buroni, S. (2017). Burkholderia cenocepacia infections in cystic fibrosis patients: drug resistance and therapeutic approaches. Front Microbiol. 8, 1592. doi: 10.3389/fmicb.2017.01592

PubMed Abstract | CrossRef Full Text | Google Scholar

Suputtamongkol, Y., Hall, A. J., Dance, D. A. B., Chaowagul, W., Rajchanuvong, A., Smith, M. D., et al. (1994). The epidemiology of melioidosis in Ubon Ratchatani, Northeast Thailand. Int. J. Epidemiol. 23, 1082–1090. doi: 10.1093/ije/23.5.1082

PubMed Abstract | CrossRef Full Text | Google Scholar

Vidyalakshmi, K., Lipika, S., Vishal, S., Damodar, S., and Chakrapani, M. (2012). Emerging clinico-epidemiological trends in melioidosis: analysis of 95 cases from western coastal India. Int. J. Infect. Dis. 16, e491–e497. doi: 10.1016/j.ijid.2012.02.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: Burkholderia spp.-transmission, host-pathogen interaction, prevention and treatment, multidrug resistance, prevention

Citation: Buroni S and Samy RP (2023) Editorial: Burkholderia spp.-transmission, pathogenesis, host-pathogen interaction, prevention and treatment. Front. Microbiol. 14:1226865. doi: 10.3389/fmicb.2023.1226865

Received: 22 May 2023; Accepted: 25 May 2023;
Published: 06 June 2023.

Edited and reviewed by: Axel Cloeckaert, Institut National de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), France

Copyright © 2023 Buroni and Samy. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Silvia Buroni, silvia.buroni@unipv.it

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.