1
|
Anifowose OR, Oladosu GA, Oridupa OA. Incidence and antimicrobial susceptibility profiles of multi-drug resistant Klebsiella pneumoniae strains isolated from diseased Clarias gariepinus. Braz J Microbiol 2025:10.1007/s42770-025-01684-z. [PMID: 40314916 DOI: 10.1007/s42770-025-01684-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Klebsiella pneumoniae is associated with substantial economic losses in the fish industry and with food-borne illness in humans. This study aimed to determine the incidence and antibiogram profiles of multi-drug resistant Klebsiella pneumoniae strains isolated from diseased Clarias gariepinus. A total of 384 diseased Clarias gariepinus that showed clinical signs of red spots and hemorrhages on the pectoral fin were sampled from commercial fish farms in Lagos, Ogun, and Oyo States, Nigeria. Samples of fin, liver, kidney, and gill were taken from diseased C. gariepinus. Samples were subjected to bacteriological examinations and isolates were identified based on morphological characteristics, biochemical tests, and 16 S rRNA gene characterisation. The retrieved isolates were tested for sequence analysis and antibiogram profile. The incidence of K. pneumoniae among the examined diseased Clarias gariepinus was 61.7% (237/384). The highest prevalence was observed in the gill (32.9%). The percentage of K. Pneumoniae isolates in diseased C. gariepinus from selected Southwest states were; Lagos State 46.8%, Ogun State 78.1%, and Oyo State 60.1% (p > 0.05). The phylogenetic analyses revealed that the tested K. pneumoniae strains shared high genetic similarity with other K. pneumoniae strains from South Africa, Saudi Arabia, India, USA, Spain, Bangladesh, and China. Antibiogram profiles revealed that 100% (120/120) of the retrieved K. pneumoniae strains were MDR to four different antimicrobial classes (penicillins, tetracyclines, nitroimidazole, and macrolides). This is the first report to reveal the occurrence of MDR K. pneumoniae in diseased C. gariepinus, an emergence that represents a risk to public health.
Collapse
Affiliation(s)
| | - Gbolahanmi Akinola Oladosu
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olayinka Ayotunde Oridupa
- Department of Veterinary Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
2
|
Shah AA, Alwashmi ASS, Abalkhail A, Alkahtani AM. Emerging challenges in Klebsiella pneumoniae: Antimicrobial resistance and novel approach. Microb Pathog 2025; 202:107399. [PMID: 39983881 DOI: 10.1016/j.micpath.2025.107399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/15/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Klebsiella pneumoniae poses significant global health challenges due to its increasing antimicrobial resistance (AMR). The emergence of multidrug-resistant (MDR) and carbapenem-resistant strains has limited treatment options, making infections difficult to control. This pathogen utilizes several mechanisms, including extended-spectrum β-lactamase (ESBL) production and efflux pumps, contributing to its resistance. Novel therapeutic approaches, such as bacteriophage therapy, CRISPR-Cas systems, and antimicrobial peptides, are being explored to combat AMR. Additionally, targeting virulence factors and biofilm formation holds promise for developing alternative strategies, providing a new frontier in tackling this formidable pathogen.
Collapse
Affiliation(s)
- Assar Ali Shah
- Department of Animal and Poultry Production, Faculty of Veterinary and Animal Science, Veterinary and Animal Science, Gomal University, Dera Ismail Khan, Pakistan; Tropical Feed Resources Research and Development Center (TROFREC), Department of Animal Science, Faculty of Agriculture, Khon Kaen University, Khon Kaen, 40002, Thailand.
| | - Ameen S S Alwashmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, 51452, Qassim, Saudi Arabia
| | - Adil Abalkhail
- Department of Public Health, College of Applied Medical Sciences, Qassim University, Buraydah, 66666, Qassim, Saudi Arabia
| | - Abdullah M Alkahtani
- Department of Microbiology & Clinical Parasitology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
3
|
Birgani AH, Goli HR, Siadat SD, Fateh A, Nikbin VS, Sakhaee F, Ashrafian F, Haghshenas MR, Gholami M. Virulence genes, efflux pumps, and molecular typing of Klebsiella pneumoniae isolates from North Iran. AMB Express 2025; 15:36. [PMID: 40045145 PMCID: PMC11883068 DOI: 10.1186/s13568-025-01845-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/14/2025] [Indexed: 03/09/2025] Open
Abstract
Resistant and virulent strains of Klebsiella pneumoniae (K. pneumoniae) are rapidly disseminated among both hospitalized patients and communities, therefore, the identification of the genes responsible for virulence and resistance, along with the clonal relatedness of these strains, could be beneficial in the management of the dissemination of these strains among patients. The aim of the present study was to assess antibiotic susceptibility, virulence and resistance genes, as well as the genetic relatedness of K. pneumoniae strains isolated from patients admitted to four hospitals in Mazandaran, Iran. A total of 95 K. pneumoniae were gathered from hospitalized patients. All isolates were confirmed using biochemical and conventional microbiological methods, followed by the assessment of susceptibility patterns through disk diffusion and the detection of resistance and virulence genes using conventional PCR. The genetic diversity of clinical isolates was determined using the Enterobacterial Repetitive Intergenic Consensus PCR (ERIC-PCR) technique. The resistance frequencies varied, with the highest being for ampicillin/sulbactam (95.8%) and the lowest for fosfomycin (3.2%). Only one strain displayed a non-MDR profile against all antibiotics tested. Virulence-associated genes were detected, such as mrkD (90.5%), fimH (80%), entB (92.6%), iutA (25.3%), and ybtS (68.4%). Genes associated with efflux pumps and outer membrane porins included acrAB (98.9%), tolC (95.8%), mdtK (83.2%), ompK35 (95.8%), and ompK36 (92.6%). Based on ERIC-PCR patterns with a 90% similarity, the isolates were categorized into 17 distinct clusters. While the majority of isolates had a same profile and were grouped in the predominant pattern, 11 isolates were identified as singletons. Our study indicates that the prevalence of MDR K. pneumoniae carrying virulence genes and exhibiting close relatedness underscores the urgent need for effective strategies to control and prevent infections caused by K. pneumoniae.
Collapse
Affiliation(s)
- Arta Hafezi Birgani
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamid Reza Goli
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Abolfazl Fateh
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | - Fatemeh Sakhaee
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Ashrafian
- Department of Clinical Research, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Reza Haghshenas
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehrdad Gholami
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
- Antimicrobial Resistance Research Center, Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
4
|
Klaper K, Pfeifer Y, Heinrich L, Prax M, Krut O, Bekeredjian-Ding I, Wahl A, Fischer MA, Kaspar H, Borgmann S, Gerlach RG, Werner G. Enhanced invasion and survival of antibiotic- resistant Klebsiella pneumoniae pathotypes in host cells and strain-specific replication in blood. Front Cell Infect Microbiol 2025; 15:1522573. [PMID: 40028183 PMCID: PMC11868097 DOI: 10.3389/fcimb.2025.1522573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
Background Klebsiella pneumoniae is one of the most important opportunistic pathogens causing healthcare-associated and community-acquired infections worldwide. In recent years, the increase in antibiotic resistance and infections caused by hypervirulent K. pneumoniae poses great public health concerns. In this study, host-pathogen interactions of different K. pneumoniae strains of human and animal origins were analyzed in microbiological, cell-biological and immunological experiments. Methods In vitro infection experiments using representatives of different K. pneumoniae pathotypes and various epithelial and macrophage cell lines were executed analyzing adhesion, invasion and intracellular replication. Experimental conditions involved normoxia and hypoxia. Furthermore, survival and growth of further K. pneumoniae isolates expressing defined siderophores in blood (platelet concentrates, serum) was investigated. All experiments were done in triplicate and statistically significant differences were determined. Results Significant differences in adhesion and invasion capability, phagocytosis resistance and intracellular replication were measured between different K. pneumoniae pathotypes. Especially, ESBL-producing K. pneumoniae isolates demonstrated increased invasion in host cell lines and survival in macrophages. A strong cytotoxic effect on intestinal cells was observed for hypervirulent K. pneumoniae. The results from our investigations of the growth behavior of K. pneumoniae in platelets and serum showed that siderophores and/or an enlarged capsule are not essential factors for the proliferation of (hypervirulent) K. pneumoniae strains in blood components. Conclusion Our in vitro experiments revealed new insights into the host-pathogen interactions of K. pneumoniae strains representing different pathovars and clonal lineages in different infectious contexts and hosts. While a clear limitation of our study is the limited strain set used for both infection and as potential host, the results are a further step for a better understanding of the pathogenicity of K. pneumoniae and its properties essential for different stages of colonization and infection. When developed further, these results may offer novel approaches for future therapeutics including novel "anti-virulence strategies".
Collapse
Affiliation(s)
- Kathleen Klaper
- Department of Infectious Diseases, Robert Koch Institute, Wernigerode, Germany
- Department of Infectious Diseases, Robert Koch Institute, Berlin, Germany
| | - Yvonne Pfeifer
- Department of Infectious Diseases, Robert Koch Institute, Wernigerode, Germany
| | - Lena Heinrich
- Department of Infectious Diseases, Robert Koch Institute, Wernigerode, Germany
| | - Marcel Prax
- Division of Microbiology, Paul Ehrlich Institute, Langen, Germany
| | - Oleg Krut
- Division of Microbiology, Paul Ehrlich Institute, Langen, Germany
| | | | - Anika Wahl
- Department of Infectious Diseases, Robert Koch Institute, Wernigerode, Germany
| | - Martin A. Fischer
- Department of Infectious Diseases, Robert Koch Institute, Wernigerode, Germany
| | - Heike Kaspar
- Division of Antibiotic Resistance Monitoring, Federal Office of Consumer Protection and Food Safety, Berlin, Germany
| | - Stefan Borgmann
- Department of Infectious Diseases and Infection Control, Hospital Ingolstadt, Ingolstadt, Germany
| | - Roman G. Gerlach
- Department of Infectious Diseases, Robert Koch Institute, Wernigerode, Germany
| | - Guido Werner
- Department of Infectious Diseases, Robert Koch Institute, Wernigerode, Germany
| |
Collapse
|
5
|
Dang J, Yu X, Zhang M, Dang L, Li Z, Shu J. Role of glycosylation in bacterial resistance to carbapenems. World J Microbiol Biotechnol 2025; 41:55. [PMID: 39883281 DOI: 10.1007/s11274-025-04272-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
Carbapenems are a class of β-lactam antibacterial drugs with a broad antibacterial spectrum and strong activity, commonly used to treat serious bacterial infections. However, improper or excessive use of carbapenems can lead to increased bacterial resistance, which is a significant concern as they are often used as last resort for treating multidrug-resistant (MDR) gram-negative bacteria. Confronted with this challenge, it is crucial to comprehensively understand the mechanism of carbapenem resistance to develop effective therapeutic strategies and innovative drugs. In recent years, emerging research on the glycosylation of bacterial proteins has highlighted the crucial role of glycans in various bacterial processes, including carbapenem resistance. Given the limited understanding of bacterial glycosylation, its role in in carbapenem resistance may be more pivotal than currently acknowledged. In this review, we summarize the direct and multifunctional role of glycosylation in bacterial resistance as well as the classical and recently reported mechanisms of bacterial carbapenem resistance, focusing on illuminating the potential role of glycosylation in carbapenem resistance. We also discuss the potential of leveraging this knowledge to develop more effective strategies for combating clinically resistant bacteria.
Collapse
Affiliation(s)
- Jing Dang
- School of Medicine, Faculty of Life Science & Medicine, Northwest University, Xi'an, China
- Laboratory for Functional Glycomics, Faculty of Life Science & Medicine, Northwest University, Xi'an, China
| | - Xinlu Yu
- School of Medicine, Faculty of Life Science & Medicine, Northwest University, Xi'an, China
| | - Mengyuan Zhang
- Laboratory for Functional Glycomics, Faculty of Life Science & Medicine, Northwest University, Xi'an, China
| | - Liuyi Dang
- Laboratory for Functional Glycomics, Faculty of Life Science & Medicine, Northwest University, Xi'an, China
| | - Zheng Li
- Laboratory for Functional Glycomics, Faculty of Life Science & Medicine, Northwest University, Xi'an, China
| | - Jian Shu
- School of Medicine, Faculty of Life Science & Medicine, Northwest University, Xi'an, China.
- Laboratory for Functional Glycomics, Faculty of Life Science & Medicine, Northwest University, Xi'an, China.
- School of Medicine, Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai Beilu, Xi'an, Shaanxi, 710069, China.
| |
Collapse
|
6
|
Xu L, Li J, Wu W, Wu X, Ren J. Klebsiella pneumoniae capsular polysaccharide: Mechanism in regulation of synthesis, virulence, and pathogenicity. Virulence 2024; 15:2439509. [PMID: 39668724 PMCID: PMC11649230 DOI: 10.1080/21505594.2024.2439509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/04/2024] [Accepted: 12/03/2024] [Indexed: 12/14/2024] Open
Abstract
Hypervirulent Klebsiella pneumoniae exhibits strong pathogenicity and can cause severe invasive infections but is historically recognized as antibiotic-susceptible. In recent years, the escalating global prevalence of antibiotic-resistant hypervirulent K. pneumoniae has raised substantial concerns and created an urgent demand for effective treatment options. Capsular polysaccharide (CPS) is one of the main virulence determinants contributing to the hypervirulent phenotype. The structure of CPS varies widely among strains, and both the structure and composition of CPS can influence the virulence of K. pneumoniae. CPS possesses various immune evasion mechanisms that promote the survival of K. pneumoniae, as well as its colonization and dissemination. Given the proven viability of therapies that target the capsule, improving our understanding of the CPS structure is critical to effectively directing treatment strategies. In this review, the structure and typing of CPS are addressed as well as genes related to synthesis and regulation, relationships with virulence, and pathogenic mechanisms. We aim to provide a reference for research on the pathogenesis of K. pneumoniae.
Collapse
Affiliation(s)
- Li Xu
- Research Institute of General Surgery, Jinling Hospital, the Affiliated Hospital of Medical School, Nanjing Medical University, Nanjing, China
| | - Jiayang Li
- Research Institute of General Surgery, Jinling Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wenqi Wu
- Research Institute of General Surgery, Jinling Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiuwen Wu
- Research Institute of General Surgery, Jinling Hospital, the Affiliated Hospital of Medical School, Nanjing Medical University, Nanjing, China
- Research Institute of General Surgery, Jinling Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, the Affiliated Hospital of Medical School, Nanjing Medical University, Nanjing, China
- Research Institute of General Surgery, Jinling Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
7
|
Abe R, Ram-Mohan N, Zudock EJ, Lewis S, Carroll KC, Yang S. Host heterogeneity in humoral bactericidal activity can be complement independent. Front Immunol 2024; 15:1457174. [PMID: 39359730 PMCID: PMC11445025 DOI: 10.3389/fimmu.2024.1457174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 08/30/2024] [Indexed: 10/04/2024] Open
Abstract
Background Humoral bactericidal activity was first recognized nearly a century ago. However, the extent of inter-individual heterogeneity and the mechanisms underlying such heterogeneity beyond antibody or complement systems have not been well studied. Methods The plasma bactericidal activity of five healthy volunteers were tested against 30 strains of Gram-negative uropathogens, Klebsiella pneumoniae and Escherichia coli, associated with bloodstream infections. IgG and IgM titers specific to K. pneumoniae strains KP13883 and KPB1 were measured by ELISA, and complement inhibitor was used to measure the contribution of complement-induced killing. Furthermore, MALDI-TOF mass spectrometry was conducted to determine the metabolomic components of plasma with bactericidal properties in 25 healthy individuals using Bayesian inference of Pearson correlation between peak intensity and colony counts of surviving bacteria. Results Plasma bactericidal activity varied widely between individuals against various bacterial strains. While individual plasma with higher IgM titers specific to K. pneumoniae strain KP13883 showed more efficient killing of the strain, both IgM and IgG titers for K. pneumoniae strain KPB1 did not correlate well with the killing activity. Complement inhibition assays elucidated that the complement-mediated killing was not responsible for the inter-individual heterogeneity in either isolate. Subsequently, using MALDI-TOF mass spectrometry on plasmas of 25 healthy individuals, we identified several small molecules including gangliosides, pediocins, or saponins as candidates that showed negative correlation between peak intensities and colony forming units of the test bacteria. Conclusion This is the first study to demonstrate the inter-individual heterogeneity of constitutive innate humoral bactericidal function quantitatively and that the heterogeneity can be independent of antibody or the complement system.
Collapse
Affiliation(s)
- Ryuichiro Abe
- Department of Emergency Medicine, Stanford University School of Medicine, Palo Alto, CA, United States
- Laboratory of Bacterial Pathogenesis, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Nikhil Ram-Mohan
- Department of Emergency Medicine, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Elizabeth Jordan Zudock
- Department of Emergency Medicine, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Shawna Lewis
- Division of Medical Microbiology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Karen C Carroll
- Division of Medical Microbiology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Samuel Yang
- Department of Emergency Medicine, Stanford University School of Medicine, Palo Alto, CA, United States
| |
Collapse
|
8
|
Goh KJ, Altuvia Y, Argaman L, Raz Y, Bar A, Lithgow T, Margalit H, Gan YH. RIL-seq reveals extensive involvement of small RNAs in virulence and capsule regulation in hypervirulent Klebsiella pneumoniae. Nucleic Acids Res 2024; 52:9119-9138. [PMID: 38804271 PMCID: PMC11347178 DOI: 10.1093/nar/gkae440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 04/29/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
Hypervirulent Klebsiella pneumoniae (hvKp) can infect healthy individuals, in contrast to classical strains that commonly cause nosocomial infections. The recent convergence of hypervirulence with carbapenem-resistance in K. pneumoniae can potentially create 'superbugs' that are challenging to treat. Understanding virulence regulation of hvKp is thus critical. Accumulating evidence suggest that posttranscriptional regulation by small RNAs (sRNAs) plays a role in bacterial virulence, but it has hardly been studied in K. pneumoniae. We applied RIL-seq to a prototypical clinical isolate of hvKp to unravel the Hfq-dependent RNA-RNA interaction (RRI) network. The RRI network is dominated by sRNAs, including predicted novel sRNAs, three of which we validated experimentally. We constructed a stringent subnetwork composed of RRIs that involve at least one hvKp virulence-associated gene and identified the capsule gene loci as a hub target where multiple sRNAs interact. We found that the sRNA OmrB suppressed both capsule production and hypermucoviscosity when overexpressed. Furthermore, OmrB base-pairs within kvrA coding region and partially suppresses translation of the capsule regulator KvrA. This agrees with current understanding of capsule as a major virulence and fitness factor. It emphasizes the intricate regulatory control of bacterial phenotypes by sRNAs, particularly of genes critical to bacterial physiology and virulence.
Collapse
Affiliation(s)
- Kwok Jian Goh
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Infection and Immunity Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Yael Altuvia
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Liron Argaman
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Yair Raz
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Amir Bar
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Trevor Lithgow
- Infection and Immunity Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Hanah Margalit
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Yunn-Hwen Gan
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| |
Collapse
|
9
|
Muner JJ, de Oliveira PAA, Baboghlian J, Moura SC, de Andrade AG, de Oliveira MM, Campos YFD, Mançano ASF, Siqueira NMG, Pacheco T, Ferraz LFC. The transcriptional regulator Fur modulates the expression of uge, a gene essential for the core lipopolysaccharide biosynthesis in Klebsiella pneumoniae. BMC Microbiol 2024; 24:279. [PMID: 39061004 PMCID: PMC11282780 DOI: 10.1186/s12866-024-03418-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Klebsiella pneumoniae is a Gram-negative pathogen that has become a threat to public health worldwide due to the emergence of hypervirulent and multidrug-resistant strains. Cell-surface components, such as polysaccharide capsules, fimbriae, and lipopolysaccharides (LPS), are among the major virulence factors for K. pneumoniae. One of the genes involved in LPS biosynthesis is the uge gene, which encodes the uridine diphosphate galacturonate 4-epimerase enzyme. Although essential for the LPS formation in K. pneumoniae, little is known about the mechanisms that regulate the expression of uge. Ferric uptake regulator (Fur) is an iron-responsive transcription factor that modulates the expression of capsular and fimbrial genes, but its role in LPS expression has not yet been identified. This work aimed to investigate the role of the Fur regulator in the expression of the K. pneumoniae uge gene and to determine whether the production of LPS by K. pneumoniae is modulated by the iron levels available to the bacterium. RESULTS Using bioinformatic analyses, a Fur-binding site was identified on the promoter region of the uge gene; this binding site was validated experimentally through Fur Titration Assay (FURTA) and DNA Electrophoretic Mobility Shift Assay (EMSA) techniques. RT-qPCR analyses were used to evaluate the expression of uge according to the iron levels available to the bacterium. The iron-rich condition led to a down-regulation of uge, while the iron-restricted condition resulted in up-regulation. In addition, LPS was extracted and quantified on K. pneumoniae cells subjected to iron-replete and iron-limited conditions. The iron-limited condition increased the amount of LPS produced by K. pneumoniae. Finally, the expression levels of uge and the amount of the LPS were evaluated on a K. pneumoniae strain mutant for the fur gene. Compared to the wild-type, the strain with the fur gene knocked out presented a lower LPS amount and an unchanged expression of uge, regardless of the iron levels. CONCLUSIONS Here, we show that iron deprivation led the K. pneumoniae cells to produce higher amount of LPS and that the Fur regulator modulates the expression of uge, a gene essential for LPS biosynthesis. Thus, our results indicate that iron availability modulates the LPS biosynthesis in K. pneumoniae through a Fur-dependent mechanism.
Collapse
Affiliation(s)
- José Júlio Muner
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, SP, Brazil
| | - Paloma Aparecida Alves de Oliveira
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, SP, Brazil
- Central Multiusuária de Análises Genômica e Transcriptômica (CmAGT), Universidade São Francisco, Bragança Paulista, SP, Brazil
| | - Juliana Baboghlian
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, SP, Brazil
| | - Stefany Casarin Moura
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, SP, Brazil
| | | | | | - Yasmin Ferreira de Campos
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, SP, Brazil
| | | | | | - Thaisy Pacheco
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, SP, Brazil
| | - Lúcio Fábio Caldas Ferraz
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, SP, Brazil.
- Central Multiusuária de Análises Genômica e Transcriptômica (CmAGT), Universidade São Francisco, Bragança Paulista, SP, Brazil.
| |
Collapse
|
10
|
Bain W, Ahn B, Peñaloza HF, McElheny CL, Tolman N, van der Geest R, Gonzalez-Ferrer S, Chen N, An X, Hosuru R, Tabary M, Papke E, Kohli N, Farooq N, Bachman W, Olonisakin TF, Xiong Z, Griffith MP, Sullivan M, Franks J, Mustapha MM, Iovleva A, Suber T, Shanks RQ, Ferreira VP, Stolz DB, Van Tyne D, Doi Y, Lee JS. In Vivo Evolution of a Klebsiella pneumoniae Capsule Defect With wcaJ Mutation Promotes Complement-Mediated Opsonophagocytosis During Recurrent Infection. J Infect Dis 2024; 230:209-220. [PMID: 39052750 PMCID: PMC11272070 DOI: 10.1093/infdis/jiae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/17/2023] [Accepted: 01/03/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Klebsiella pneumoniae carbapenemase-producing K pneumoniae (KPC-Kp) bloodstream infections are associated with high mortality. We studied clinical bloodstream KPC-Kp isolates to investigate mechanisms of resistance to complement, a key host defense against bloodstream infection. METHODS We tested growth of KPC-Kp isolates in human serum. In serial isolates from a single patient, we performed whole genome sequencing and tested for complement resistance and binding by mixing study, direct enzyme-linked immunosorbent assay, flow cytometry, and electron microscopy. We utilized an isogenic deletion mutant in phagocytosis assays and an acute lung infection model. RESULTS We found serum resistance in 16 of 59 (27%) KPC-Kp clinical bloodstream isolates. In 5 genetically related bloodstream isolates from a single patient, we noted a loss-of-function mutation in the capsule biosynthesis gene, wcaJ. Disruption of wcaJ was associated with decreased polysaccharide capsule, resistance to complement-mediated killing, and surprisingly, increased binding of complement proteins. Furthermore, an isogenic wcaJ deletion mutant exhibited increased opsonophagocytosis in vitro and impaired in vivo control in the lung after airspace macrophage depletion in mice. CONCLUSIONS Loss of function in wcaJ led to increased complement resistance, complement binding, and opsonophagocytosis, which may promote KPC-Kp persistence by enabling coexistence of increased bloodstream fitness and reduced tissue virulence.
Collapse
Affiliation(s)
- William Bain
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Brian Ahn
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus School of Medicine, Denver
| | - Hernán F Peñaloza
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | | | - Nathanial Tolman
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Rick van der Geest
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Shekina Gonzalez-Ferrer
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Nathalie Chen
- Division of Infectious Diseases, Department of Medicine
| | - Xiaojing An
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Ria Hosuru
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Mohammadreza Tabary
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Erin Papke
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Naina Kohli
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | | | | | - Tolani F Olonisakin
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Zeyu Xiong
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | | | - Mara Sullivan
- Center for Biologic Imaging, Department of Cell Biology
| | | | | | - Alina Iovleva
- Division of Infectious Diseases, Department of Medicine
| | - Tomeka Suber
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Robert Q Shanks
- Department of Ophthalmology, University of Pittsburgh, Pennsylvania
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Ohio
| | - Donna B Stolz
- Center for Biologic Imaging, Department of Cell Biology
| | | | - Yohei Doi
- Division of Infectious Diseases, Department of Medicine
| | - Janet S Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
- Division of Pulmonary and Critical Care Medicine, Washington University in St Louis, Missouri
| |
Collapse
|
11
|
Saberi Riseh R, Gholizadeh Vazvani M, Taheri A, Kennedy JF. Pectin-associated immune responses in plant-microbe interactions: A review. Int J Biol Macromol 2024; 273:132790. [PMID: 38823736 DOI: 10.1016/j.ijbiomac.2024.132790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/04/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
This review explores the role of pectin, a complex polysaccharide found in the plant cell wall, in mediating immune responses during interactions between plants and microbes. The objectives of this study were to investigate the molecular mechanisms underlying pectin-mediated immune responses and to understand how these interactions shape plant-microbe communication. Pectin acts as a signaling molecule, triggering immune responses such as the production of antimicrobial compounds, reinforcement of the cell wall, and activation of defense-related genes. Pectin functions as a target for pathogen-derived enzymes, enabling successful colonization by certain microbial species. The document discusses the complexity of pectin-based immune signaling networks and their modulation by various factors, including pathogen effectors and host proteins. It also emphasizes the importance of understanding the crosstalk between pectin-mediated immunity and other defense pathways to develop strategies for enhancing plant resistance against diseases. The insights gained from this study have implications for the development of innovative approaches to enhance crop protection and disease management in agriculture. Further investigations into the components and mechanisms involved in pectin-mediated immunity will pave the way for future advancements in plant-microbe interaction research.
Collapse
Affiliation(s)
- Roohallah Saberi Riseh
- Department of Plant Protection, Faculty of Agriculture, Vali-e-Asr University of Rafsanjan, 7718897111 Rafsanjan, Iran.
| | - Mozhgan Gholizadeh Vazvani
- Department of Plant Protection, Faculty of Agriculture, Vali-e-Asr University of Rafsanjan, 7718897111 Rafsanjan, Iran
| | - Abdolhossein Taheri
- Department of Plant Protection, Faculty of Plant Production, University of agricultural Sciences and natural resources of Gorgan, Iran.
| | - John F Kennedy
- Chembiotech Laboratories Ltd, WR15 8FF Tenbury Wells, United Kingdom.
| |
Collapse
|
12
|
Raju M, Parasuraman B, Govindasamy P, Thangavelu P, Duraisamy S. Improved anti-diabetic and anticancer activities of green synthesized CuO nanoparticles derived from Tabernaemontana divaricate leaf extract. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:46498-46512. [PMID: 36905548 DOI: 10.1007/s11356-023-26261-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
Copper oxide nanoparticles (CuO NPs) are among the most commonly employed nanoparticle materials owing to their antibacterial qualities, although their primary mechanism of action (MOA) is still not completely understood. CuO NPs are synthesized in this study using leaf extract of Tabernaemontana divaricate (TDCO3), and they are then analyzed using XRD, FT-IR, SEM, and EDX analysis. The zone of inhibition of TDCO3 NPs against both gram-positive (G+) B. subtilis and gram-negative (G-) K. pneumoniae bacteria was 34 mm and 33 mm, respectively. Furthermore, Cu2+/Cu+ ions promote reactive oxygen species and electrostatically bind with the negatively charged teichoic acid in the bacterial cell wall. The anti-inflammatory and anti-diabetics analysis was conducted using standard BSA denaturation and α-amylase inhibition technique with cell inhibition values of 85.66 and 81.18% for TDCO3 NPs. Additionally, the TDCO3 NPs delivered prominent anticancer activity with the lowest IC50 value 18.2 μg/mL in the MTT assay technique against HeLa cancer cells.
Collapse
Affiliation(s)
- Manonmani Raju
- PG and Research Department of Physics, Arignar Anna Government Arts College, Namakkal, Tamil Nadu, 637002, India
| | - Balaji Parasuraman
- Smart Materials Laboratory, Department of Physics, Periyar University, Salem, Tamil Nadu, 636011, India
| | - Palanisamy Govindasamy
- Smart Materials Laboratory, Department of Physics, Periyar University, Salem, Tamil Nadu, 636011, India
| | - Pazhanivel Thangavelu
- Smart Materials Laboratory, Department of Physics, Periyar University, Salem, Tamil Nadu, 636011, India
| | - Sasikumar Duraisamy
- PG and Research Department of Physics, Arignar Anna Government Arts College, Namakkal, Tamil Nadu, 637002, India.
| |
Collapse
|
13
|
Lê-Bury P, Echenique-Rivera H, Pizarro-Cerdá J, Dussurget O. Determinants of bacterial survival and proliferation in blood. FEMS Microbiol Rev 2024; 48:fuae013. [PMID: 38734892 PMCID: PMC11163986 DOI: 10.1093/femsre/fuae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 05/13/2024] Open
Abstract
Bloodstream infection is a major public health concern associated with high mortality and high healthcare costs worldwide. Bacteremia can trigger fatal sepsis whose prevention, diagnosis, and management have been recognized as a global health priority by the World Health Organization. Additionally, infection control is increasingly threatened by antimicrobial resistance, which is the focus of global action plans in the framework of a One Health response. In-depth knowledge of the infection process is needed to develop efficient preventive and therapeutic measures. The pathogenesis of bloodstream infection is a dynamic process resulting from the invasion of the vascular system by bacteria, which finely regulate their metabolic pathways and virulence factors to overcome the blood immune defenses and proliferate. In this review, we highlight our current understanding of determinants of bacterial survival and proliferation in the bloodstream and discuss their interactions with the molecular and cellular components of blood.
Collapse
Affiliation(s)
- Pierre Lê-Bury
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Autoimmune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 18 route du Panorama, 92260 Fontenay-aux-Roses, France
| | - Hebert Echenique-Rivera
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France
| | - Javier Pizarro-Cerdá
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, Yersinia National Reference Laboratory, WHO Collaborating Research & Reference Centre for Plague FRA-146, 28 rue du Dr Roux, 75015 Paris, France
| | - Olivier Dussurget
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Yersinia Research Unit, 28 rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
14
|
Bai R, Guo J. Interactions and Implications of Klebsiella pneumoniae with Human Immune Responses and Metabolic Pathways: A Comprehensive Review. Infect Drug Resist 2024; 17:449-462. [PMID: 38333568 PMCID: PMC10849896 DOI: 10.2147/idr.s451013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
Klebsiella pneumoniae (K. pneumoniae), a significant contributor to the global challenge of antibiotic resistance, is not only a ubiquitous component of the human microbiome but also a potent pathogen capable of causing a spectrum of diseases. This review provides a thorough analysis of the intricate interactions between K. pneumoniae and the human immune system, elucidating its substantial impact on metabolic processes. We explore the mechanisms employed by K. pneumoniae to evade and manipulate immune responses, including molecular mimicry, immune modulation, and biofilm formation. The review further investigates the bacterium's influence on metabolic pathways, particularly glycolysis, highlighting how these interactions exacerbate disease severity. The emergence of multidrug-resistant and extremely drug-resistant strains within the Enterobacteriaceae family has heightened the public health crisis, underscoring the urgency for comprehensive research. We investigate the roles of the host's complement system, autophagy, cell death mechanisms, and various cytokines in combating K. pneumoniae infections, shedding light on areas that warrant further academic investigation. Additionally, the review discusses the challenges posed by K1- and K2-capsule polysaccharides in vaccine development due to their complex molecular structures and adhesive properties. Acknowledging the limited availability of effective antimicrobials, this review advocates for exploring alternative approaches such as immunotherapeutics, vaccinations, and phage therapy. We consolidate current knowledge on K. pneumoniae, covering classical and non-classical subtypes, antimicrobial resistance-mediated genes, virulence factors, and epidemiological trends in isolation and antibiotic resistance rates. This comprehensive review not only advances our understanding of K. pneumoniae but also underscores the imperative for ongoing research and collaborative efforts to develop new prevention and treatment strategies against this formidable pathogen.
Collapse
Affiliation(s)
- Ruojing Bai
- Department of Geriatric Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, People’s Republic of China
| | - Jun Guo
- Department of Geriatric Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, People’s Republic of China
| |
Collapse
|
15
|
Behera LM, Ghosh M, Gupta PK, Rana S. A rationally engineered small antimicrobial peptide with potent antibacterial activity. J Cell Biochem 2024; 125:e30503. [PMID: 37992185 DOI: 10.1002/jcb.30503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023]
Abstract
Antimicrobial resistance (AMR) is a silent pandemic declared by the WHO that requires urgent attention in the post-COVID world. AMR is a critical public health concern worldwide, potentially affecting people at different stages of life, including the veterinary and agriculture industries. Notably, very few new-age antimicrobial agents are in the current developmental pipeline. Thus, the design, discovery, and development of new antimicrobial agents are required to address the menace of AMR. Antimicrobial peptides (AMPs) are an important class of antimicrobial agents for combating AMR due to their broad-spectrum activity and ability to evade AMR through a multimodal mechanism of action. However, molecular size, aggregability, proteolytic degradation, cytotoxicity, and hemolysis activity significantly limit the clinical application of natural AMPs. The de novo design and engineering of a short synthetic amphipathic AMP (≤16 aa, Mol. Wt. ≤ 2 kDa) with an unusual architecture comprised of coded and noncoded amino acids (NCAAs) is presented here, which demonstrates potent antibacterial activity against a few selected bacterial strains mentioned in the WHO priority list. The designer AMP is conformationally ordered in solution and effectively permeabilizes the outer and inner membranes, leading to bacterial growth inhibition and death. Additionally, the peptide is resistant to proteolysis and has negligible cytotoxicity and hemolysis activity up to 150 μM toward cultured human cell lines and erythrocytes. The designer AMP is unique and appears to be a potent therapeutic candidate, which can be subsequently subjected to preclinical studies to explicitly understand and address the menace of AMR.
Collapse
Affiliation(s)
- Lalita Mohan Behera
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Manaswini Ghosh
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Pulkit Kr Gupta
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Soumendra Rana
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| |
Collapse
|
16
|
Sebola DC, Oguttu JW, Kock MM, Qekwana DN. Antimicrobial resistance patterns of Acinetobacter baumannii and Klebsiella pneumoniae isolated from dogs presented at a veterinary academic hospital in South Africa. Vet World 2023; 16:1880-1888. [PMID: 37859969 PMCID: PMC10583888 DOI: 10.14202/vetworld.2023.1880-1888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 08/22/2023] [Indexed: 10/21/2023] Open
Abstract
Background Acinetobacter baumannii and Klebsiella pneumoniae are opportunistic bacterial pathogens responsible for hospital-acquired infections in veterinary medicine. Infection with these bacteria always requires urgent antimicrobial therapy. However, there is no evidence of studies that have investigated the antimicrobial drug resistance profile of these organisms in a veterinary setting in South Africa. This study investigated the antimicrobial resistance (AMR) patterns of A. baumannii and K. pneumoniae from clinical specimens obtained from dogs presented at a veterinary academic hospital. The findings of this study contribute to an improved understanding of the AMR profile of these bacteria in veterinary medicine. Materials and Methods Retrospective data of clinical samples from dogs that were positive for A. baumannii and K. pneumoniae between 2007 and 2013 were used in this study. The antimicrobial susceptibility of the isolates was determined using the disk diffusion method following the Clinical and Laboratory Standards Institute guidelines. The A. baumannii isolates were subjected to a panel of 20 antibiotics, while K. pneumoniae isolates were subjected to a panel of 22 antibiotics. Data were analyzed using descriptive statistics and presented using tables and figures. Results Twenty (n = 20) A. baumannii isolates were isolated from bronchoalveolar lavage, foreign objects, bone, urine, skin, blood, ear, nasal, and oral cavity. Almost all A. baumannii (95%, 19/20) isolates were resistant to at least one antibiotic, and 60% (12/20) were multidrug-resistant (MDR). Klebsiella pneumoniae (n = 56) was isolated from urine, foreign objects, abscesses, ears, eyes, tracheal aspirations, bronchoalveolar lavages, eyes, abdominal aspirates, anal glands, bones, and intestinal and lung biopsies. All K. pneumoniae (100%, 56/56) isolates were resistant to at least one antibiotic, and 98% (55/56) were MDR. Conclusion Both A. baumannii and K. pneumoniae were isolated in various clinical tissue samples and exhibited a high prevalence of resistance to multiple antibiotics. In addition, these bacteria exhibited a high prevalence of resistance to β-lactam compared to other classes of antibiotics, which is likely to impact treatment options and patient prognosis.
Collapse
Affiliation(s)
- Dikeledi C. Sebola
- Section Veterinary Public Health, Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - James W. Oguttu
- Department of Agriculture and Animal Health, College of Agriculture and Environmental Sciences, University of South Africa, Johannesburg, South Africa
| | - Marleen M. Kock
- Department of Medical Microbiology, University of Pretoria, Pretoria, South Africa
- Tshwane Academic Division, National Health Laboratory Service, Pretoria, South Africa
| | - Daniel N. Qekwana
- Section Veterinary Public Health, Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
17
|
Jati AP, Sola-Campoy PJ, Bosch T, Schouls LM, Hendrickx APA, Bautista V, Lara N, Raangs E, Aracil B, Rossen JWA, Friedrich AW, Navarro Riaza AM, Cañada-García JE, Ramírez de Arellano E, Oteo-Iglesias J, Pérez-Vázquez M, García-Cobos S. Widespread Detection of Yersiniabactin Gene Cluster and Its Encoding Integrative Conjugative Elements (ICE Kp) among Nonoutbreak OXA-48-Producing Klebsiella pneumoniae Clinical Isolates from Spain and the Netherlands. Microbiol Spectr 2023; 11:e0471622. [PMID: 37310221 PMCID: PMC10434048 DOI: 10.1128/spectrum.04716-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 05/22/2023] [Indexed: 06/14/2023] Open
Abstract
In this study, we determined the presence of virulence factors in nonoutbreak, high-risk clones and other isolates belonging to less common sequence types associated with the spread of OXA-48-producing Klebsiella pneumoniae clinical isolates from The Netherlands (n = 61) and Spain (n = 53). Most isolates shared a chromosomally encoded core of virulence factors, including the enterobactin gene cluster, fimbrial fim and mrk gene clusters, and urea metabolism genes (ureAD). We observed a high diversity of K-Locus and K/O loci combinations, KL17 and KL24 (both 16%), and the O1/O2v1 locus (51%) being the most prevalent in our study. The most prevalent accessory virulence factor was the yersiniabactin gene cluster (66.7%). We found seven yersiniabactin lineages-ybt 9, ybt 10, ybt 13, ybt 14, ybt 16, ybt 17, and ybt 27-which were chromosomally embedded in seven integrative conjugative elements (ICEKp): ICEKp3, ICEKp4, ICEKp2, ICEKp5, ICEKp12, ICEKp10, and ICEKp22, respectively. Multidrug-resistant lineages-ST11, ST101, and ST405-were associated with ybt 10/ICEKp4, ybt 9/ICEKp3, and ybt 27/ICEKp22, respectively. The fimbrial adhesin kpi operon (kpiABCDEFG) was predominant among ST14, ST15, and ST405 isolates, as well as the ferric uptake system kfuABC, which was also predominant among ST101 isolates. No convergence of hypervirulence and resistance was observed in this collection of OXA-48-producing K. pneumoniae clinical isolates. Nevertheless, two isolates, ST133 and ST792, were positive for the genotoxin colibactin gene cluster (ICEKp10). In this study, the integrative conjugative element, ICEKp, was the major vehicle for yersiniabactin and colibactin gene clusters spreading. IMPORTANCE Convergence of multidrug resistance and hypervirulence in Klebsiella pneumoniae isolates has been reported mostly related to sporadic cases or small outbreaks. Nevertheless, little is known about the real prevalence of carbapenem-resistant hypervirulent K. pneumoniae since these two phenomena are often separately studied. In this study, we gathered information on the virulent content of nonoutbreak, high-risk clones (i.e., ST11, ST15, and ST405) and other less common STs associated with the spread of OXA-48-producing K. pneumoniae clinical isolates. The study of virulence content in nonoutbreak isolates can help us to expand information on the genomic landscape of virulence factors in K. pneumoniae population by identifying virulence markers and their mechanisms of spread. Surveillance should focus not only on antimicrobial resistance but also on virulence characteristics to avoid the spread of multidrug and (hyper)virulent K. pneumoniae that may cause untreatable and more severe infections.
Collapse
Affiliation(s)
- Afif P. Jati
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
- Indonesian Society of Bioinformatics and Biodiversity, Indonesia
| | - Pedro J. Sola-Campoy
- Laboratorio de Referencia e Investigación en Resistencia a Antibióticos e Infecciones Relacionadas con la Asistencia Sanitaria, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Thijs Bosch
- Infectious Diseases Research, Diagnostics and Laboratory Surveillance, Centre for Infectious Disease Control Netherlands, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Leo M. Schouls
- Infectious Diseases Research, Diagnostics and Laboratory Surveillance, Centre for Infectious Disease Control Netherlands, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Antoni P. A. Hendrickx
- Infectious Diseases Research, Diagnostics and Laboratory Surveillance, Centre for Infectious Disease Control Netherlands, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Verónica Bautista
- Laboratorio de Referencia e Investigación en Resistencia a Antibióticos e Infecciones Relacionadas con la Asistencia Sanitaria, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Noelia Lara
- Laboratorio de Referencia e Investigación en Resistencia a Antibióticos e Infecciones Relacionadas con la Asistencia Sanitaria, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Erwin Raangs
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
| | - Belén Aracil
- Laboratorio de Referencia e Investigación en Resistencia a Antibióticos e Infecciones Relacionadas con la Asistencia Sanitaria, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- CIBER de Enfermedades Infecciosas, Spanish Network for Research in Infectious Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - John W. A. Rossen
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
- Laboratory of Medical Microbiology and Infectious Diseases, Isala Hospital, Zwolle, The Netherlands
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Alex W. Friedrich
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
- University Hospital Münster, Institute of European Prevention Networks in Infection Control, Münster, Germany
| | - Ana M. Navarro Riaza
- Laboratorio de Referencia e Investigación en Resistencia a Antibióticos e Infecciones Relacionadas con la Asistencia Sanitaria, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Javier E. Cañada-García
- Laboratorio de Referencia e Investigación en Resistencia a Antibióticos e Infecciones Relacionadas con la Asistencia Sanitaria, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Eva Ramírez de Arellano
- Laboratorio de Referencia e Investigación en Resistencia a Antibióticos e Infecciones Relacionadas con la Asistencia Sanitaria, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- CIBER de Enfermedades Infecciosas, Spanish Network for Research in Infectious Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Oteo-Iglesias
- Laboratorio de Referencia e Investigación en Resistencia a Antibióticos e Infecciones Relacionadas con la Asistencia Sanitaria, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- CIBER de Enfermedades Infecciosas, Spanish Network for Research in Infectious Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - María Pérez-Vázquez
- Laboratorio de Referencia e Investigación en Resistencia a Antibióticos e Infecciones Relacionadas con la Asistencia Sanitaria, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- CIBER de Enfermedades Infecciosas, Spanish Network for Research in Infectious Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Silvia García-Cobos
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
- Laboratorio de Referencia e Investigación en Resistencia a Antibióticos e Infecciones Relacionadas con la Asistencia Sanitaria, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - The Dutch and Spanish Collaborative Working Groups on Surveillance on Carbapenemase-Producing Enterobacterales
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection Prevention, Groningen, The Netherlands
- Indonesian Society of Bioinformatics and Biodiversity, Indonesia
- Laboratorio de Referencia e Investigación en Resistencia a Antibióticos e Infecciones Relacionadas con la Asistencia Sanitaria, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Infectious Diseases Research, Diagnostics and Laboratory Surveillance, Centre for Infectious Disease Control Netherlands, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- CIBER de Enfermedades Infecciosas, Spanish Network for Research in Infectious Diseases, Instituto de Salud Carlos III, Madrid, Spain
- Laboratory of Medical Microbiology and Infectious Diseases, Isala Hospital, Zwolle, The Netherlands
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
- University Hospital Münster, Institute of European Prevention Networks in Infection Control, Münster, Germany
| |
Collapse
|
18
|
Wunrow HY, Bender RG, Vongpradith A, Sirota SB, Swetschinski LR, Novotney A, Gray AP, Ikuta KS, Sharara F, Wool EE, Aali A, Abd-Elsalam S, Abdollahi A, Abdul Aziz JM, Abidi H, Aboagye RG, Abolhassani H, Abu-Gharbieh E, Adamu LH, Adane TD, Addo IY, Adegboye OA, Adekiya TA, Adnan M, Adnani QES, Afzal S, Aghamiri S, Aghdam ZB, Agodi A, Ahinkorah BO, Ahmad A, Ahmad S, Ahmadzade M, Ahmed A, Ahmed A, Ahmed JQ, Ahmed MS, Akinosoglou K, Aklilu A, Akonde M, Alahdab F, AL-Ahdal TMA, Alanezi FM, Albelbeisi AH, Alemayehu TBB, Alene KA, Al-Eyadhy A, Al-Gheethi AAS, Ali A, Ali BA, Ali L, Ali SS, Alimohamadi Y, Alipour V, Aljunid SM, Almustanyir S, Al-Raddadi RM, Alvis-Guzman N, Al-Worafi YM, Aly H, Ameyaw EK, Ancuceanu R, Ansar A, Ansari G, Anyasodor AE, Arabloo J, Aravkin AY, Areda D, Artamonov AA, Arulappan J, Aruleba RT, Asaduzzaman M, Atalell KA, Athari SS, Atlaw D, Atout MMW, Attia S, Awoke T, Ayalew MK, Ayana TM, Ayele AD, Azadnajafabad S, Azizian K, Badar M, Badiye AD, Baghcheghi N, Bagheri M, Bagherieh S, Bahadory S, Baig AA, Barac A, Barati S, Bardhan M, Basharat Z, Bashiri A, Basnyat B, Bassat Q, Basu S, Bayileyegn NS, Bedi N, et alWunrow HY, Bender RG, Vongpradith A, Sirota SB, Swetschinski LR, Novotney A, Gray AP, Ikuta KS, Sharara F, Wool EE, Aali A, Abd-Elsalam S, Abdollahi A, Abdul Aziz JM, Abidi H, Aboagye RG, Abolhassani H, Abu-Gharbieh E, Adamu LH, Adane TD, Addo IY, Adegboye OA, Adekiya TA, Adnan M, Adnani QES, Afzal S, Aghamiri S, Aghdam ZB, Agodi A, Ahinkorah BO, Ahmad A, Ahmad S, Ahmadzade M, Ahmed A, Ahmed A, Ahmed JQ, Ahmed MS, Akinosoglou K, Aklilu A, Akonde M, Alahdab F, AL-Ahdal TMA, Alanezi FM, Albelbeisi AH, Alemayehu TBB, Alene KA, Al-Eyadhy A, Al-Gheethi AAS, Ali A, Ali BA, Ali L, Ali SS, Alimohamadi Y, Alipour V, Aljunid SM, Almustanyir S, Al-Raddadi RM, Alvis-Guzman N, Al-Worafi YM, Aly H, Ameyaw EK, Ancuceanu R, Ansar A, Ansari G, Anyasodor AE, Arabloo J, Aravkin AY, Areda D, Artamonov AA, Arulappan J, Aruleba RT, Asaduzzaman M, Atalell KA, Athari SS, Atlaw D, Atout MMW, Attia S, Awoke T, Ayalew MK, Ayana TM, Ayele AD, Azadnajafabad S, Azizian K, Badar M, Badiye AD, Baghcheghi N, Bagheri M, Bagherieh S, Bahadory S, Baig AA, Barac A, Barati S, Bardhan M, Basharat Z, Bashiri A, Basnyat B, Bassat Q, Basu S, Bayileyegn NS, Bedi N, Behnoush AH, Bekel AA, Belete MA, Bello OO, Bhagavathula AS, Bhandari D, Bhardwaj P, Bhaskar S, Bhat AN, Bijani A, Bineshfar N, Boloor A, Bouaoud S, Buonsenso D, Burkart K, Cámera LA, Castañeda-Orjuela CA, Cernigliaro A, Charan J, Chattu VK, Ching PR, Chopra H, Choudhari SG, Christopher DJ, Chu DT, Couto RAS, Cruz-Martins N, Dadras O, Dai X, Dandona L, Dandona R, Das S, Dash NR, Dashti M, De la Hoz FP, Debela SA, Dejen D, Dejene H, Demeke D, Demeke FM, Demessa BH, Demetriades AK, Demissie S, Dereje D, Dervišević E, Desai HD, Dessie AM, Desta F, Dhama K, Djalalinia S, Do TC, Dodangeh M, Dodangeh M, Dominguez RMV, Dongarwar D, Dsouza HL, Durojaiye OC, Dziedzic AM, Ekat MH, Ekholuenetale M, Ekundayo TC, El Sayed Zaki M, El-Abid H, Elhadi M, El-Hajj VG, El-Huneidi W, El-Sakka AA, Esayas HL, Fagbamigbe AF, Falahi S, Fares J, Fatehizadeh A, Fatima SAF, Feasey NA, Fekadu G, Fetensa G, Feyissa D, Fischer F, Foroutan B, Gaal PA, Gadanya MA, Gaipov A, Ganesan B, Gebrehiwot M, Gebrekidan KG, Gebremeskel TG, Gedef GM, Gela YY, Gerema U, Gessner BD, Getachew ME, Ghadiri K, Ghaffari K, Ghamari SH, Ghanbari R, Ghazy RMM, Ghozali G, Gizaw ABAB, Glushkova EV, Goldust M, Golechha M, Guadie HA, Guled RA, Gupta M, Gupta S, Gupta VB, Gupta VK, Gupta VK, Hadi NR, Haj-Mirzaian A, Haller S, Hamidi S, Haque S, Harapan H, Hasaballah AI, Hasan I, Hasani H, Hasanian M, Hassankhani H, Hassen MB, Hayat K, Heidari M, Heidari-Foroozan M, Heidari-Soureshjani R, Hezam K, Holla R, Horita N, Hossain MM, Hosseini MS, Hosseinzadeh M, Hostiuc S, Hussain S, Hussein NR, Ibitoye SE, Ilesanmi OS, Ilic IM, Ilic MD, Imam MT, Iregbu KC, Ismail NE, Iwu CCD, Jaja C, Jakovljevic M, Jamshidi E, Javadi Mamaghani A, Javidnia J, Jokar M, Jomehzadeh N, Joseph N, Joshua CE, Jozwiak JJ, Kabir Z, Kalankesh LR, Kalhor R, Kamal VK, Kandel H, Karaye IM, Karch A, Karimi H, Kaur H, Kaur N, Keykhaei M, Khajuria H, Khalaji A, Khan A, Khan IA, Khan M, Khan T, Khatab K, Khatatbeh MM, Khayat Kashani HR, Khubchandani J, Kim MS, Kisa A, Kisa S, Kompani F, Koohestani HR, Kothari N, Krishan K, Krishnamoorthy Y, Kulimbet M, Kumar M, Kumaran SD, Kuttikkattu A, Kwarteng A, Laksono T, Landires I, Laryea DO, Lawal BK, Le TTT, Ledda C, Lee SW, Lee S, Lema GK, Levi M, Lim SS, Liu X, Lopes G, Lutzky Saute R, Machado Teixeira PH, Mahmoodpoor A, Mahmoud MA, Malakan Rad E, Malhotra K, Malik AA, Martinez-Guerra BA, Martorell M, Mathur V, Mayeli M, Medina JRC, Melese A, Memish ZA, Mentis AFA, Merza MA, Mestrovic T, Michalek IM, Minh LHN, Mirahmadi A, Mirmosayyeb O, Misganaw A, Misra AK, Moghadasi J, Mohamed NS, Mohammad Y, Mohammadi E, Mohammed S, Mojarrad Sani M, Mojiri-forushani H, Mokdad AH, Momtazmanesh S, Monasta L, Moni MA, Mossialos E, Mostafavi E, Motaghinejad M, Mousavi Khaneghah A, Mubarik S, Muccioli L, Muhammad JS, Mulita F, Mulugeta T, Murillo-Zamora E, Mustafa G, Muthupandian S, Nagarajan AJ, Nainu F, Nair TS, Nargus S, Nassereldine H, Natto ZS, Nayak BP, Negoi I, Negoi RI, Nejadghaderi SA, Nguyen HQ, Nguyen PT, Nguyen VT, Niazi RK, Noroozi N, Nouraei H, Nuñez-Samudio V, Nuruzzaman KM, Nwatah VE, Nzoputam CI, Nzoputam OJ, Oancea B, Obaidur RM, Odetokun IA, Ogunsakin RE, Okonji OC, Olagunju AT, Olana LT, Olufadewa II, Oluwafemi YD, Oumer KS, Ouyahia A, P A M, Pakshir K, Palange PN, Pardhan S, Parikh RR, Patel J, Patel UK, Patil S, Paudel U, Pawar S, Pensato U, Perdigão J, Pereira M, Peres MFP, Petcu IR, Pinheiro M, Piracha ZZ, Pokhrel N, Postma MJ, Prates EJS, Qattea I, Raghav PR, Rahbarnia L, Rahimi-Movaghar V, Rahman M, Rahman MA, Rahmanian V, Rahnavard N, Ramadan H, Ramasubramani P, Rani U, Rao IR, Rapaka D, Ratan ZA, Rawaf S, Redwan EMM, Reiner Jr RC, Rezaei N, Riad A, Ribeiro da Silva TM, Roberts T, Robles Aguilar G, Rodriguez JAB, Rosenthal VD, Saddik B, Sadeghian S, Saeed U, Safary A, Saheb Sharif-Askari F, Saheb Sharif-Askari N, Sahebkar A, Sahu M, Sajedi SA, Saki M, Salahi S, Salahi S, Saleh MA, Sallam M, Samadzadeh S, Samy AM, Sanjeev RK, Satpathy M, Seylani A, Sha'aban A, Shafie M, Shah PA, Shahrokhi S, Shahzamani K, Shaikh MA, Sham S, Shannawaz M, Sheikh A, Shenoy SM, Shetty PH, Shin JI, Shokri F, Shorofi SA, Shrestha S, Sibhat MM, Siddig EE, Silva LMLR, Singh H, Singh JA, Singh P, Singh S, Sinto R, Skryabina AA, Socea B, Sokhan A, Solanki R, Solomon Y, Sood P, Soshnikov S, Stergachis A, Sufiyan MB, Suliankatchi Abdulkader R, Sultana A, T Y SS, Taheri E, Taki E, Tamuzi JJLL, Tan KK, Tat NY, Temsah MH, Terefa DR, Thangaraju P, Tibebu NS, Ticoalu JHV, Tillawi T, Tincho MB, Tleyjeh II, Toghroli R, Tovani-Palone MR, Tufa DG, Turner P, Ullah I, Umeokonkwo CD, Unnikrishnan B, Vahabi SM, Vaithinathan AG, Valizadeh R, Varthya SB, Vos T, Waheed Y, Walde MT, Wang C, Weerakoon KG, Wickramasinghe ND, Winkler AS, Woldemariam M, Worku NA, Wright C, Yada DY, Yaghoubi S, Yahya GATY, Yenew CYY, Yesiltepe M, Yi S, Yiğit V, You Y, Yusuf H, Zakham F, Zaman M, Zaman SB, Zare I, Zareshahrabadi Z, Zarrintan A, Zastrozhin MS, Zhang H, Zhang J, Zhang ZJ, Zheng P, Zoladl M, Zumla A, Hay SI, Murray CJL, Naghavi M, Kyu HH. Global, regional, and national burden of meningitis and its aetiologies, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Neurol 2023; 22:685-711. [PMID: 37479374 PMCID: PMC10356620 DOI: 10.1016/s1474-4422(23)00195-3] [Show More Authors] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/28/2023] [Accepted: 05/05/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Although meningitis is largely preventable, it still causes hundreds of thousands of deaths globally each year. WHO set ambitious goals to reduce meningitis cases by 2030, and assessing trends in the global meningitis burden can help track progress and identify gaps in achieving these goals. Using data from the Global Burden of Diseases, Injuries, and Risk Factors Study (GBD) 2019, we aimed to assess incident cases and deaths due to acute infectious meningitis by aetiology and age from 1990 to 2019, for 204 countries and territories. METHODS We modelled meningitis mortality using vital registration, verbal autopsy, sample-based vital registration, and mortality surveillance data. Meningitis morbidity was modelled with a Bayesian compartmental model, using data from the published literature identified by a systematic review, as well as surveillance data, inpatient hospital admissions, health insurance claims, and cause-specific meningitis mortality estimates. For aetiology estimation, data from multiple causes of death, vital registration, hospital discharge, microbial laboratory, and literature studies were analysed by use of a network analysis model to estimate the proportion of meningitis deaths and cases attributable to the following aetiologies: Neisseria meningitidis, Streptococcus pneumoniae, Haemophilus influenzae, group B Streptococcus, Escherichia coli, Klebsiella pneumoniae, Listeria monocytogenes, Staphylococcus aureus, viruses, and a residual other pathogen category. FINDINGS In 2019, there were an estimated 236 000 deaths (95% uncertainty interval [UI] 204 000-277 000) and 2·51 million (2·11-2·99) incident cases due to meningitis globally. The burden was greatest in children younger than 5 years, with 112 000 deaths (87 400-145 000) and 1·28 million incident cases (0·947-1·71) in 2019. Age-standardised mortality rates decreased from 7·5 (6·6-8·4) per 100 000 population in 1990 to 3·3 (2·8-3·9) per 100 000 population in 2019. The highest proportion of total all-age meningitis deaths in 2019 was attributable to S pneumoniae (18·1% [17·1-19·2]), followed by N meningitidis (13·6% [12·7-14·4]) and K pneumoniae (12·2% [10·2-14·3]). Between 1990 and 2019, H influenzae showed the largest reduction in the number of deaths among children younger than 5 years (76·5% [69·5-81·8]), followed by N meningitidis (72·3% [64·4-78·5]) and viruses (58·2% [47·1-67·3]). INTERPRETATION Substantial progress has been made in reducing meningitis mortality over the past three decades. However, more meningitis-related deaths might be prevented by quickly scaling up immunisation and expanding access to health services. Further reduction in the global meningitis burden should be possible through low-cost multivalent vaccines, increased access to accurate and rapid diagnostic assays, enhanced surveillance, and early treatment. FUNDING Bill & Melinda Gates Foundation.
Collapse
|
19
|
St. John A, Perault AI, Giacometti SI, Sommerfield AG, DuMont AL, Lacey KA, Zheng X, Sproch J, Petzold C, Dancel-Manning K, Gonzalez S, Annavajhala M, Beckford C, Zeitouni N, Liang FX, van Bakel H, Shopsin B, Uhlemann AC, Pironti A, Torres VJ. Capsular Polysaccharide Is Essential for the Virulence of the Antimicrobial-Resistant Pathogen Enterobacter hormaechei. mBio 2023; 14:e0259022. [PMID: 36779722 PMCID: PMC10127600 DOI: 10.1128/mbio.02590-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/13/2023] [Indexed: 02/14/2023] Open
Abstract
Nosocomial infections caused by multidrug-resistant (MDR) Enterobacter cloacae complex (ECC) pathogens are on the rise. However, the virulence strategies employed by these pathogens remain elusive. Here, we study the interaction of ECC clinical isolates with human serum to define how this pathogen evades the antimicrobial action of complement, one of the first lines of host-mediated immune defense. We identified a small number of serum-sensitive strains, including Enterobacter hormaechei strain NR3055, which we exploited for the in vitro selection of serum-resistant clones. Comparative genomics between the serum-sensitive NR3055 strain and the isolated serum-resistant clones revealed a premature stop codon in the wzy gene of the capsular polysaccharide biosynthesis locus of NR3055. The complementation of wzy conferred serum resistance to NR3055, prevented the deposition of complement proteins on the bacterial surface, inhibited phagocytosis by human neutrophils, and rendered the bacteria virulent in a mouse model of peritonitis. Mice exposed to a nonlethal dose of encapsulated NR3055 were protected from subsequent lethal infections by encapsulated NR3055, whereas mice that were previously exposed to unencapsulated NR3055 succumbed to infection. Thus, capsule is a key immune evasion determinant for E. hormaechei, and it is a potential target for prophylactics and therapeutics to combat these increasingly MDR human pathogens. IMPORTANCE Infections caused by antimicrobial resistant bacteria are of increasing concern, especially those due to carbapenem-resistant Enterobacteriaceae pathogens. Included in this group are species of the Enterobacter cloacae complex, regarding which there is a paucity of knowledge on the infection biology of the pathogens, despite their clinical relevance. In this study, we combine techniques in comparative genomics, bacterial genetics, and diverse models of infection to establish capsule as an important mechanism of Enterobacter pathogens to resist the antibacterial activity of serum, a first line of host defense against bacterial infections. We also show that immune memory targeting the Enterobacter capsule protects against lethal infection. The further characterization of Enterobacter infection biology and the immune response to infection are needed for the development of therapies and preventative interventions targeting these highly antibiotic resistant pathogens.
Collapse
Affiliation(s)
- Amelia St. John
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
| | - Andrew I. Perault
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
| | - Sabrina I. Giacometti
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| | - Alexis G. Sommerfield
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Ashley L. DuMont
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Keenan A. Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Xuhui Zheng
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Julia Sproch
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Chris Petzold
- Microscopy Laboratory, Division of Advanced Research Technologies, New York University Langone Health, New York, New York, USA
| | - Kristen Dancel-Manning
- Microscopy Laboratory, Division of Advanced Research Technologies, New York University Langone Health, New York, New York, USA
| | - Sandra Gonzalez
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Medini Annavajhala
- Department of Medicine, Division of Infectious Diseases, Columbia University Medical Center, New York, New York, USA
| | - Colleen Beckford
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nathalie Zeitouni
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Feng-Xia Liang
- Microscopy Laboratory, Division of Advanced Research Technologies, New York University Langone Health, New York, New York, USA
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, New York, USA
| | - Anne-Catrin Uhlemann
- Department of Medicine, Division of Infectious Diseases, Columbia University Medical Center, New York, New York, USA
| | - Alejandro Pironti
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Microbial Computational Genomic Core Lab, Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
20
|
Gelfusa M, Murari A, Ludovici GM, Franchi C, Gelfusa C, Malizia A, Gaudio P, Farinelli G, Panella G, Gargiulo C, Casinelli K. On the Potential of Relational Databases for the Detection of Clusters of Infection and Antibiotic Resistance Patterns. Antibiotics (Basel) 2023; 12:antibiotics12040784. [PMID: 37107146 PMCID: PMC10135313 DOI: 10.3390/antibiotics12040784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
In recent years, several bacterial strains have acquired significant antibiotic resistance and can, therefore, become difficult to contain. To counteract such trends, relational databases can be a powerful tool for supporting the decision-making process. The case of Klebsiella pneumoniae diffusion in a central region of Italy was analyzed as a case study. A specific relational database is shown to provide very detailed and timely information about the spatial-temporal diffusion of the contagion, together with a clear assessment of the multidrug resistance of the strains. The analysis is particularized for both internal and external patients. Tools such as the one proposed can, therefore, be considered important elements in the identification of infection hotspots, a key ingredient of any strategy to reduce the diffusion of an infectious disease at the community level and in hospitals. These types of tools are also very valuable in the decision-making process related to antibiotic prescription and to the management of stockpiles. The application of this processing technology to viral diseases such as COVID-19 is under investigation.
Collapse
Affiliation(s)
- Michela Gelfusa
- Department of Industrial Engineering, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Andrea Murari
- Consorzio RFX (CNR, ENEA, INFN), University of Padua, 35127 Padua, Italy
- Istituto per la Scienza e la Tecnologia dei Plasmi, CNR, 35100 Padua, Italy
| | - Gian Marco Ludovici
- Department of Industrial Engineering, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Cristiano Franchi
- Department of Industrial Engineering, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Claudio Gelfusa
- Department of Industrial Engineering, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Andrea Malizia
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Pasqualino Gaudio
- Department of Industrial Engineering, University of Rome "Tor Vergata", 00133 Rome, Italy
| | | | - Giacinto Panella
- ASL and Fabrizio Spaziani, Frosinone Hospital, 03100 Frosinone, Italy
| | - Carla Gargiulo
- ASL and Fabrizio Spaziani, Frosinone Hospital, 03100 Frosinone, Italy
| | - Katia Casinelli
- ASL and Fabrizio Spaziani, Frosinone Hospital, 03100 Frosinone, Italy
| |
Collapse
|
21
|
Tohda M, Oinuma KI, Sakiyama A, Tsubouchi T, Niki M, Namikawa H, Yamane K, Yamada K, Watanabe T, Asai K, Kakeya H, Kaneko Y, Kawaguchi T. Rifampicin exerts anti-mucoviscous activity against hypervirulent Klebsiella pneumoniae via binding to the RNA polymerase β subunit. J Glob Antimicrob Resist 2023; 32:21-28. [PMID: 36572148 DOI: 10.1016/j.jgar.2022.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVES In hypervirulent Klebsiella pneumoniae (hvKP), the hypermucoviscous capsule is known to be a major virulence determinant. We previously discovered that rifampicin (RFP), a bactericidal drug that binds to and inhibits the β subunit of RNA polymerase (RpoB), elicits anti-mucoviscous activity against hvKP by suppressing rmpA, a regulator of capsule production. Here, we aimed to determine whether RFP exerts this effect at sub-growth-inhibitory concentrations via its binding to RpoB. METHODS Five spontaneous RFP-resistant mutants (R1-R5) were prepared from an hvKP clinical isolate and subjected to whole genome sequencing and mucoviscosity analyses. Subsequently, a two-step allelic exchange procedure was used to create a rpoB mutant R6 and revertants with wild-type rpoB from R1-R5 (named R1'-R5'). Transcription levels of rmpA and the capsular polysaccharide polymerase gene magA and capsule thickness of R1-R5 and R1'-R5' grown without or with RFP were evaluated by quantitative reverse transcription polymerase chain reaction and microscopic observation using India ink staining. RESULTS R1-R5 all had non-synonymous point mutations in rpoB and were highly resistant to the bactericidal effects and anti-mucoviscous activity of RFP. While the properties of R6 were similar to those of R1-R5, the responses of R1'-R5' to RFP were identical to those of the wild type. rmpA and magA transcription levels and capsule thickness correlated well with the mucoviscosity levels. CONCLUSIONS RFP exerts anti-mucoviscous activity by binding to RpoB. The mechanism of how this causes rmpA suppression remains to be explored.
Collapse
Affiliation(s)
- Mitsunori Tohda
- Department of Respiratory Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Ken-Ichi Oinuma
- Department of Bacteriology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan; Research Center for Infectious Disease Sciences, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan.
| | - Arata Sakiyama
- Department of Bacteriology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Taishi Tsubouchi
- Department of Bacteriology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan; Research Center for Infectious Disease Sciences, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Mamiko Niki
- Department of Bacteriology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan; Research Center for Infectious Disease Sciences, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hiroki Namikawa
- Department of Medical Education and General Practice, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Kenshi Yamane
- Department of Respiratory Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Koichi Yamada
- Research Center for Infectious Disease Sciences, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan; Department of Infection Control Science, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Tetsuya Watanabe
- Department of Respiratory Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Kazuhisa Asai
- Department of Respiratory Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Kakeya
- Research Center for Infectious Disease Sciences, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan; Department of Infection Control Science, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yukihiro Kaneko
- Department of Bacteriology, Osaka Metropolitan University, Graduate School of Medicine, Osaka, Japan; Research Center for Infectious Disease Sciences, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Tomoya Kawaguchi
- Department of Respiratory Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
22
|
Carbapenem-Resistant Klebsiella pneumoniae: Virulence Factors, Molecular Epidemiology and Latest Updates in Treatment Options. Antibiotics (Basel) 2023; 12:antibiotics12020234. [PMID: 36830145 PMCID: PMC9952820 DOI: 10.3390/antibiotics12020234] [Citation(s) in RCA: 112] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023] Open
Abstract
Klebsiella pneumoniae is a Gram-negative opportunistic pathogen responsible for a variety of community and hospital infections. Infections caused by carbapenem-resistant K. pneumoniae (CRKP) constitute a major threat for public health and are strongly associated with high rates of mortality, especially in immunocompromised and critically ill patients. Adhesive fimbriae, capsule, lipopolysaccharide (LPS), and siderophores or iron carriers constitute the main virulence factors which contribute to the pathogenicity of K. pneumoniae. Colistin and tigecycline constitute some of the last resorts for the treatment of CRKP infections. Carbapenemase production, especially K. pneumoniae carbapenemase (KPC) and metallo-β-lactamase (MBL), constitutes the basic molecular mechanism of CRKP emergence. Knowledge of the mechanism of CRKP appearance is crucial, as it can determine the selection of the most suitable antimicrobial agent among those most recently launched. Plazomicin, eravacycline, cefiderocol, temocillin, ceftolozane-tazobactam, imipenem-cilastatin/relebactam, meropenem-vaborbactam, ceftazidime-avibactam and aztreonam-avibactam constitute potent alternatives for treating CRKP infections. The aim of the current review is to highlight the virulence factors and molecular pathogenesis of CRKP and provide recent updates on the molecular epidemiology and antimicrobial treatment options.
Collapse
|
23
|
Opstrup KV, Christiansen G, Birkelund S. Beta-lactam induced morphological changes in serum of extended-spectrum beta-lactamase-producing Klebsiella pneumoniae blood isolates. Microbes Infect 2023; 25:105036. [PMID: 35944888 DOI: 10.1016/j.micinf.2022.105036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 07/15/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023]
Abstract
Klebsiella pneumoniae is an opportunistic pathogen, which frequently causes bacteremia. Ceftazidime and meropenem, two important beta-lactam antibiotics for treatment of K. pneumoniae infections, induce morphological changes in bacteria when examined in vitro. Thirty clinical Klebsiella spp. Bacteremia isolates were analyzed for antimicrobial resistance and serum resistance. To determine whether complement influenced the resistance to ceftazidime of extended-spectrum beta-lactamase producing-isolates and sensitivity to meropenem, one serum resistant and one partly serum sensitive isolate were analyzed in normal human serum, heat-inactivated human serum, and growth medium with addition of beta-lactam antibiotics. HA391 was resistant to ceftazidime and had identical minimum inhibitory concentrations for meropenem in normal human serum, heat-inactivated serum and RPMI. In normal human serum, HA233 was inhibited by ceftazidime and had lower inhibitory concentrations of meropenem. Morphological changes induced by serum and beta-lactam antibiotics were analyzed by light- and electron microscopy. Light microscopy showed elongation of bacteria treated with ceftazidime. By electron microscopy membrane attack complexes were observed for HA233 in normal human serum, thereby facilitating beta-lactam antibiotics access to the periplasmic space and the peptidoglycan layer, explaining the increased killing of HA233 by beta-lactam antibiotics. Complement did not enhance beta-lactam killing of HA391, underlining the importance of serum susceptibility.
Collapse
Affiliation(s)
- Katharina V Opstrup
- Department of Health Science and Technology, Medical Microbiology and Immunology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg, Denmark.
| | - Gunna Christiansen
- Department of Health Science and Technology, Medical Microbiology and Immunology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg, Denmark.
| | - Svend Birkelund
- Department of Health Science and Technology, Medical Microbiology and Immunology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg, Denmark.
| |
Collapse
|
24
|
Tang L, Wang H, Cao K, Li Y, Li T, Huang Y, Xu Y. Epidemiological Features and Impact of High Glucose Level on Virulence Gene Expression and Serum Resistance of Klebsiella pneumoniae Causing Liver Abscess in Diabetic Patients. Infect Drug Resist 2023; 16:1221-1230. [PMID: 36879852 PMCID: PMC9985391 DOI: 10.2147/idr.s391349] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/18/2023] [Indexed: 03/05/2023] Open
Abstract
Purpose Klebsiella pneumoniae (K. pneumoniae) is a Gram-negative bacterium that is predominantly associated with liver abscesses in global diabetic patients. High levels of glucose in the surrounding of K. pneumonia increase its pathogenicity including capsular polysaccharide (CPS) and fimbriae. Other important virulent factors include outer membrane protein A (ompA) and regulator mucoid phenotype A (rmpA). The objective of this investigation was to elucidate the effects of high glucose on rmpA and ompA gene expression and serum resistance of K. pneumoniae causing liver abscess. Patients and Methods The clinical history of 57 patients suffering from K. pneumoniae-caused liver abscesses (KLA) was acquired and their clinical and laboratory manifestations in the presence or absence of diabetes were analyzed. The antimicrobial susceptibility, serotypes, and virulence genes were tested. Clinical isolates of 3 serotype-K1 hypervirulent K. pneumoniae (hvKP) were used to detect the effect of exogenous high glucose on rmpA, ompA, and clbB genes expression, and bacterial serum resistance. Results KLA patients with diabetes showed higher C-reactive protein (CRP) compared to non-diabetic KLA patients. Furthermore, the diabetic group showed increased incidences of sepsis and invasive infections, and their length of hospital stay was also prolonged. Pre-incubation of K. pneumoniae in high glucose (0.5%) concentration up-regulated rmpA, ompA, and clbB genes expression. However, cAMP supplementation, which was inhibited by environmental glucose, reversed the increase of rmpA and ompA in a cAMP-dependent manner. Moreover, hvKP strains incubated in high glucose also exhibited enhanced protection from serum killing. Conclusion High glucose levels reflected by poor glycemic control has increased gene expression of rmpA and ompA in hvKP by the cAMP signaling pathway and enhanced its resistance to serum killing, thus providing a new and reasonable explanation for the high incidences of sepsis and invasive infections in KLA patients with diabetes.
Collapse
Affiliation(s)
- Ling Tang
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Hui Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Kangli Cao
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Yajuan Li
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Tingting Li
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Ying Huang
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Yuanhong Xu
- Department of Clinical Laboratory, First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
25
|
Hu MY, Chen YW, Chai ZF, Wang YZ, Lin JQ, Fang SG. Antibacterial Properties and Potential Mechanism of Serum from Chinese Alligator. Microorganisms 2022; 10:2210. [PMID: 36363802 PMCID: PMC9698150 DOI: 10.3390/microorganisms10112210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 04/06/2025] Open
Abstract
The Chinese alligator (Alligator sinensis) is an ancient reptile with strong immunity that lives in wetland environments. This study tested the antibacterial ability of Chinese alligator serum (CAS) against Klebsiella pneumoniae, Escherichia coli, Staphylococcus aureus, and Pseudomonas aeruginosa and analyzed the potential underlying mechanisms. Results showed that the CAS had a marked antibacterial effect on K. pneumoniae, E. coli, and P. aeruginosa, while S. aureus was only mildly affected. However, these effects disappeared when Protease K was added to the serum. The serum proteome analysis revealed that the antibacterial ability of CAS was produced by interactions among various proteins and that the complement proteins played a major antibacterial role. Therefore, we made relevant predictions about the structure and function of complement component 3. In addition, sequence alignment and phylogenetic analysis of complement component 3d (C3d) in four mammalian species and two alligator species showed that the amino acids that make up the acid pocket on the concave surface of alligator C3d are not identical to those in mammals. This study provided evidence that CAS elicits significant antibacterial effects against some pathogens and provides the basis for further development of novel antibacterial drugs.
Collapse
Affiliation(s)
- Meng-Yuan Hu
- MOE Key Laboratory of Biosystems Homeostasis and Protection, State Conservation Center for Gene Resources of Endangered Wildlife, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yi-Wen Chen
- MOE Key Laboratory of Biosystems Homeostasis and Protection, State Conservation Center for Gene Resources of Endangered Wildlife, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhi-Fan Chai
- MOE Key Laboratory of Biosystems Homeostasis and Protection, State Conservation Center for Gene Resources of Endangered Wildlife, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yin-Zhi Wang
- MOE Key Laboratory of Biosystems Homeostasis and Protection, State Conservation Center for Gene Resources of Endangered Wildlife, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jian-Qing Lin
- Guangdong Provincial Key Laboratory of Marine Disaster Prediction and Prevention, Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Science, Shantou University, Shantou 515063, China
| | - Sheng-Guo Fang
- MOE Key Laboratory of Biosystems Homeostasis and Protection, State Conservation Center for Gene Resources of Endangered Wildlife, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
26
|
Opstrup KV, Bennike TB, Christiansen G, Birkelund S. Complement killing of clinical Klebsiella pneumoniae isolates is serum concentration dependent. Microbes Infect 2022; 25:105074. [PMID: 36336240 DOI: 10.1016/j.micinf.2022.105074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Klebsiella pneumoniae is an opportunistic gram-negative pathogen causing serious infections, including sepsis. In plasma, activation of the complement cascades is important for killing bacteria. Thirty clinical Klebsiella spp. blood isolates were analyzed for serum susceptibility in 75% normal human serum (NHS). Twenty-two were serum resistant and eight were serum sensitive, and subsequently tested in 5-75% NHS. Two isolates were killed in 5% and the remaining six in 50%-75% NHS. The two 5% sensitive isolates showed binding of complement (C)4 and C3 in 5% NHS with formation of membrane attack complex (MAC). Inhibition of the classical/lectin mediated pathways (CP/LP) using a C4 specific nanobody, hC4Nb8, led to survival of both isolates in 5% NHS. Using nanobody hC3Nb1, inhibiting the alternative pathway (AP), the isolates were killed in 5% NHS, and amplification of the CP/LP by AP was not necessary for killing. Sole AP killing of these isolates when inhibiting CP/LP with hC4Nb8 was observed in 50% NHS, stressing the concentration dependent functionality of AP. For the less sensitive isolates, killing required activation of CP/LP and AP demonstrated by inhibition with nanobodies. AP inhibition resulted in no C3 deposition on the serum resistant isolate, supporting that AP was the sole activation pathway.
Collapse
Affiliation(s)
- Katharina V Opstrup
- Department of Health Science and Technology, Medical Microbiology and Immunology, Aalborg University, Fredrik Bajers Vej 5, 9220, Aalborg East, Denmark
| | - Tue B Bennike
- Department of Health Science and Technology, Medical Microbiology and Immunology, Aalborg University, Fredrik Bajers Vej 5, 9220, Aalborg East, Denmark
| | - Gunna Christiansen
- Department of Health Science and Technology, Medical Microbiology and Immunology, Aalborg University, Fredrik Bajers Vej 5, 9220, Aalborg East, Denmark
| | - Svend Birkelund
- Department of Health Science and Technology, Medical Microbiology and Immunology, Aalborg University, Fredrik Bajers Vej 5, 9220, Aalborg East, Denmark.
| |
Collapse
|
27
|
Wei S, Xu T, Chen Y, Zhou K. Autophagy, cell death, and cytokines in K. pneumoniae infection: Therapeutic Perspectives. Emerg Microbes Infect 2022; 12:2140607. [DOI: 10.1080/22221751.2022.2140607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Sha Wei
- Shenzhen Institute of Respiratory Diseases, Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University; the First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Tingting Xu
- Shenzhen Institute of Respiratory Diseases, Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University; the First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Yuxin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, Jiangsu, China
| | - Kai Zhou
- Shenzhen Institute of Respiratory Diseases, Second Clinical Medical College (Shenzhen People’s Hospital), Jinan University; the First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
28
|
Sommer K, Kowald S, Chopra-Dewasthaly R. Serum Resistance of Mycoplasma agalactiae Strains and Mutants Bearing Different Lipoprotein Profiles. Pathogens 2022; 11:pathogens11091036. [PMID: 36145468 PMCID: PMC9501237 DOI: 10.3390/pathogens11091036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 12/05/2022] Open
Abstract
In order to spread systemically, resistance against complement and other factors present in serum is an important trait in pathogenic bacteria. The variable proteins of Mycoplasma agalactiae (Vpmas) have been shown to affect differential adhesion, invasion and immune evasion, and undergo high-frequency phase-variation in expression. However, nothing is known about their involvement in M. agalactiae’s serum susceptibility. To evaluate this, the PG2 strain, the GM139 strain and the six Vpma phase-locked mutants (PLMs, PLMU to PLMZ) were tested for their ability to survive in the presence of non-sensitized and sensitized sheep serum, as well as guinea pig complement. Additionally, the reactivity of the sensitized sheep serum was analysed on the strains via western blotting. Overall data demonstrate PG2 strain to be more susceptible to sheep serum compared to the GM139 strain bearing a different Vpma profile. Significant differences were also observed between the different PLMs, with PLMU and PLMX showing the highest serum susceptibility in serum, while the other PLMs expressing longer Vpma proteins were more resistant. The results are in good correlation with previous studies where shorter lipoprotein variants contributed to a higher susceptibility to complement. Since none of the tested strains and PLMs were susceptible to non-sensitized sheep serum, antibodies seem to play an important role in serum killing.
Collapse
|
29
|
Doorduijn DJ, Lukassen MV, van 't Wout MFL, Franc V, Ruyken M, Bardoel BW, Heck AJR, Rooijakkers SHM. Soluble MAC is primarily released from MAC-resistant bacteria that potently convert complement component C5. eLife 2022; 11:77503. [PMID: 35947526 PMCID: PMC9402229 DOI: 10.7554/elife.77503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 08/01/2022] [Indexed: 11/21/2022] Open
Abstract
The membrane attack complex (MAC or C5b-9) is an important effector of the immune system to kill invading microbes. MAC formation is initiated when complement enzymes on the bacterial surface convert complement component C5 into C5b. Although the MAC is a membrane-inserted complex, soluble forms of MAC (sMAC), or terminal complement complex (TCC), are often detected in sera of patients suffering from infections. Consequently, sMAC has been proposed as a biomarker, but it remains unclear when and how it is formed during infections. Here, we studied mechanisms of MAC formation on different Gram-negative and Gram-positive bacteria and found that sMAC is primarily formed in human serum by bacteria resistant to MAC-dependent killing. Surprisingly, C5 was converted into C5b more potently by MAC-resistant compared to MAC-sensitive Escherichia coli strains. In addition, we found that MAC precursors are released from the surface of MAC-resistant bacteria during MAC assembly. Although release of MAC precursors from bacteria induced lysis of bystander human erythrocytes, serum regulators vitronectin (Vn) and clusterin (Clu) can prevent this. Combining size exclusion chromatography with mass spectrometry profiling, we show that sMAC released from bacteria in serum is a heterogeneous mixture of complexes composed of C5b-8, up to three copies of C9 and multiple copies of Vn and Clu. Altogether, our data provide molecular insight into how sMAC is generated during bacterial infections. This fundamental knowledge could form the basis for exploring the use of sMAC as biomarker.
Collapse
Affiliation(s)
- Dennis J Doorduijn
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marie V Lukassen
- Biomolecular Mass Spectrometry and Proteomics, Utrecht University, Utrecht, Netherlands
| | - Marije F L van 't Wout
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Vojtech Franc
- Biomolecular Mass Spectrometry and Proteomics, Utrecht University, Utrecht, Netherlands
| | - Maartje Ruyken
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Bart W Bardoel
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Utrecht University, Utrecht, Netherlands
| | - Suzan H M Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
30
|
Huang X, Li X, An H, Wang J, Ding M, Wang L, Li L, Ji Q, Qu F, Wang H, Xu Y, Lu X, He Y, Zhang JR. Capsule type defines the capability of Klebsiella pneumoniae in evading Kupffer cell capture in the liver. PLoS Pathog 2022; 18:e1010693. [PMID: 35914009 PMCID: PMC9342791 DOI: 10.1371/journal.ppat.1010693] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 06/22/2022] [Indexed: 11/21/2022] Open
Abstract
Polysaccharide capsule is the main virulence factor of K. pneumoniae, a major pathogen of bloodstream infections in humans. While more than 80 capsular serotypes have been identified in K. pneumoniae, only several serotypes are frequently identified in invasive infections. It is documented that the capsule enhances bacterial resistance to phagocytosis, antimicrobial peptides and complement deposition under in vitro conditions. However, the precise role of the capsule in the process of K. pneumoniae bloodstream infections remains to be elucidated. Here we show that the capsule promotes K. pneumoniae survival in the bloodstream by protecting bacteria from being captured by liver resident macrophage Kupffer cells (KCs). Our real-time in vivo imaging revealed that blood-borne acapsular K. pneumoniae mutant is rapidly captured and killed by KCs in the liver sinusoids of mice, whereas, to various extents, encapsulated strains bypass the anti-bacterial machinery in a serotype-dependent manner. Using capsule switched strains, we show that certain high-virulence (HV) capsular serotypes completely block KC’s capture, whereas the low-virulence (LV) counterparts confer partial protection against KC’s capture. Moreover, KC’s capture of the LV K. pneumoniae could be in vivo neutralized by free capsular polysaccharides of homologous but not heterologous serotypes, indicating that KCs specifically recognize the LV capsules. Finally, immunization with inactivated K. pneumoniae enables KCs to capture the HV K. pneumoniae. Together, our findings have uncovered that KCs are the major target cells of K. pneumoniae capsule to promote bacterial survival and virulence, which can be reversed by vaccination. Klebsiella pneumoniae is a major human pathogen. While capsule is the main virulence factor of the pathogen, only several of more than 80 capsule serotypes are frequently identified in invasive infections. However, it remains unclear how capsule contributes to K. pneumoniae virulence. Here we show that capsule type defines K. pneumoniae virulence by differential escape of immune surveillance in the liver. While low-virulence (LV) types are captured by Kupffer cells (KCs), high-virulence (HV) types circumvent the anti-bacterial machinery. Further, inactivated K. pneumoniae vaccine enables KCs to capture the HV K. pneumoniae and protects mice from lethal infection. Our findings explain the clinical prevalence of HV capsule types, and provide promising insights for future vaccine development.
Collapse
Affiliation(s)
- Xueting Huang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Xiuyuan Li
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
| | - Haoran An
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Juanjuan Wang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Ming Ding
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
| | - Lijun Wang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
- Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Lulu Li
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
| | - Quanjiang Ji
- School of Physical Science and Technology, Shanghai Tech University, Shanghai, China
| | - Fen Qu
- The Center of Clinical Diagnosis Laboratory, 302 Hospital of PLA, Beijing, China
- China Aviation General Hospital of China Medical University, Beijing, China
| | - Hui Wang
- Department of Clinical Laboratory, Peking University People’s Hospital, Beijing, China
| | - Yingchun Xu
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinxin Lu
- Department of Clinical Laboratory, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yuan He
- Research Beyond Borders, Boehringer Ingelheim (China), Shanghai, China
| | - Jing-Ren Zhang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- * E-mail:
| |
Collapse
|
31
|
Pavan HK, Shreevatsa B, Dharmashekara C, Shruthi G, Prasad KS, S Patil S, Shivamallu C. Review of Known and Unknown Facts of Klebsiella Pneumoniae and its Relationship with Antibiotics. BIOMEDICAL AND PHARMACOLOGY JOURNAL 2022; 15:643-650. [DOI: 10.13005/bpj/2403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Antibiotics are commonly used to treat bacterial respiratory infections, but they can exacerbate inflammation by releasing microbial components that overstimulate the immune system, leading to greater tissue damage. Klebsiella pneumoniae is a gram-negative, rod-shaped bacteria of the family Enterobacteriaceae. Knowing about Klebsiella pneumoniae is extremely important in the present situation, as it is one of the major causal organisms of pneumonia. Internal and external factors of K. pneumoniae are responsible for the entry and multiplication inside the host. Antibiotics against K. pneumoniae are a class of Penicillins, Cephalosporins, Monobactams, and Carbapenems which have the β-lactam ring in common with variable side chains. Combating the antibiotics by synthesizing the enzymes like beta-lactamases is the main reason for the survival of these organisms against newer generation antibiotics. In this review, we have tried to discuss about Klebsiella pneumoniae, antibiotics, and their mechanism of action.
Collapse
Affiliation(s)
- Heggadadevanakote Kendaganna Pavan
- 1Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysore, Karnataka, India
| | - Bhargav Shreevatsa
- 1Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysore, Karnataka, India
| | - Chandan Dharmashekara
- 1Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysore, Karnataka, India
| | | | - Kollur Shiva Prasad
- 3Department of Sciences, Amrita School of Arts and Sciences, Amrita Vishwa Vidyapeetham, Mysuru Campus, Mysuru – 570 026, Karnataka, India
| | - Sharanagouda S Patil
- 4ICAR-National Institute of Veterinary Epidemiology and Disease Informatics (NIVEDI), Bengaluru, Karnataka, India
| | - Chandan Shivamallu
- 1Department of Biotechnology and Bioinformatics, School of Life Sciences, JSS Academy of Higher Education and Research, Mysore, Karnataka, India
| |
Collapse
|
32
|
Mohamed H, Marusich E, Afanasev Y, Leonov S. Bacterial Outer Membrane Permeability Increase Underlies the Bactericidal Effect of Fatty Acids From Hermetia illucens (Black Soldier Fly) Larvae Fat Against Hypermucoviscous Isolates of Klebsiella pneumoniae. Front Microbiol 2022; 13:844811. [PMID: 35602017 PMCID: PMC9121012 DOI: 10.3389/fmicb.2022.844811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/08/2022] [Indexed: 11/25/2022] Open
Abstract
Behind expensive treatments, Klebsiella pneumoniae infections account for extended hospitalization’s high mortality rates. This study aimed to evaluate the activity and mechanism of the antimicrobial action of a fatty acid-containing extract (AWME3) isolated from Hermetia illucens (HI) larvae fat against K. pneumoniae subsp. pneumoniae standard NDM-1 carbapenemase-producing ATCC BAA-2473 strain, along with a wild-type hypermucoviscous clinical isolate, strain K. pneumoniae subsp. pneumoniae KPi1627, and an environmental isolate, strain K. pneumoniae subsp. pneumoniae KPM9. We classified these strains as extensive multidrug-resistant (XDR) or multiple antibiotic-resistant (MDR) demonstrated by a susceptibility assay against 14 antibiotics belonging to ten classes of antibiotics. Antibacterial properties of fatty acids extracted from the HI larvae fat were evaluated using disk diffusion method, microdilution, minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), half of the inhibitory concentration (MIC50), and bactericidal assays. In addition, the cytotoxocity of AWME3 was tested on human HEK293 cells, and AWME3 lipid profile was determined by gas chromatography-mass spectrometry (GC-MS) analysis. For the first time, we demonstrated that the inhibition zone diameter (IZD) of fatty acid-containing extract (AWME3) of the HI larvae fat tested at 20 mg/ml was 16.52 ± 0.74 and 14.23 ± 0.35 mm against colistin-resistant KPi1627 and KPM9, respectively. It was 19.72 ± 0.51 mm against the colistin-susceptible K. pneumoniae ATCC BAA-2473 strain. The MIC and MBC were 250 μg/ml for all the tested bacteria strains, indicating the bactericidal effect of AWME3. The MIC50 values were 155.6 ± 0.009 and 160.1 ± 0.008 μg/ml against the KPi1627 and KPM9 isolates, respectively, and 149.5 ± 0.013 μg/ml against the ATCC BAA-2473 strain in the micro-dilution assay. For the first time, we demonstrated that AWME3 dose-dependently increased bacterial cell membrane permeability as determined by the relative electric conductivity (REC) of the K. pneumoniae ATCC BAA-2473 suspension, and that none of the strains did not build up resistance to extended AWME3 treatment using the antibiotic resistance assay. Cytotoxicity assay showed that AWME3 is safe for human HEK293 cells at IC50 266.1 μg/ml, while bactericidal for all the strains of bacteria at the same concentration. Free fatty acids (FFAs) and their derivatives were the significant substances among 33 compounds identified by the GC-MS analysis of AWME3. Cis-oleic and palmitoleic acids represent the most abundant unsaturated FAs (UFAs), while palmitic, lauric, stearic, and myristic acids were the most abundant saturated FAs (SFAs) of the AWME3 content. Bactericidal resistant-free AWM3 mechanism of action provides a rationale interpretations and the utility of HI larvae fat to develop natural biocidal resistance-free formulations that might be promising therapeutic against Gram-negative MDR bacteria causing nosocomial infections.
Collapse
Affiliation(s)
- Heakal Mohamed
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Elena Marusich
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- *Correspondence: Elena Marusich,
| | - Yuriy Afanasev
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Sergey Leonov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Institute of Cell Biophysics, Russian Academy of Sciences, Moscow, Russia
- Sergey Leonov,
| |
Collapse
|
33
|
Mende K, Akers KS, Tyner SD, Bennett JW, Simons MP, Blyth DM, Li P, Stewart L, Tribble DR. Multidrug-Resistant and Virulent Organisms Trauma Infections: Trauma Infectious Disease Outcomes Study Initiative. Mil Med 2022; 187:42-51. [PMID: 35512375 DOI: 10.1093/milmed/usab131] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION During the wars in Iraq and Afghanistan, increased incidence of multidrug-resistant (MDR) organisms, as well as polymicrobial wounds and infections, complicated the management of combat trauma-related infections. Multidrug resistance and wound microbiology are a research focus of the Trauma Infectious Disease Outcomes Study (TIDOS), an Infectious Disease Clinical Research Program, Uniformed Services University, research protocol. To conduct comprehensive microbiological research with the goal of improving the understanding of the complicated etiology of wound infections, the TIDOS MDR and Virulent Organisms Trauma Infections Initiative (MDR/VO Initiative) was established as a collaborative effort with the Brooke Army Medical Center, Naval Medical Research Center, U.S. Army Institute of Surgical Research, and Walter Reed Army Institute of Research. We provide a review of the TIDOS MDR/VO Initiative and summarize published findings. METHODS Antagonism and biofilm formation of commonly isolated wound bacteria (e.g., ESKAPE pathogens-Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.), antimicrobial susceptibility patterns, and clinical outcomes are being examined. Isolates collected from admission surveillance swabs, as part of infection control policy, and clinical infection workups were retained in the TIDOS Microbiological Repository and associated clinical data in the TIDOS database. RESULTS Over the TIDOS study period (June 2009 to December 2014), more than 8,300 colonizing and infecting isolates were collected from military personnel injured with nearly one-third of isolates classified as MDR. At admission to participating U.S. military hospitals, 12% of wounded warriors were colonized with MDR Gram-negative bacilli. Furthermore, 27% of 913 combat casualties with ≥1 infection during their trauma hospitalization had MDR Gram-negative bacterial infections. Among 335 confirmed combat-related extremity wound infections (2009-2012), 61% were polymicrobial and comprised various combinations of Gram-negative and Gram-positive bacteria, yeast, fungi, and anaerobes. Escherichia coli was the most common Gram-negative bacilli isolated from clinical workups, as well as the most common colonizing MDR secondary to extended-spectrum β-lactamase resistance. Assessment of 479 E. coli isolates collected from wounded warriors found 188 pulsed-field types (PFTs) from colonizing isolates and 54 PFTs from infecting isolates without significant overlap across combat theaters, military hospitals, and study years. A minority of patients with colonizing E. coli isolates developed subsequent infections with the same E. coli strain. Enterococcus spp. were most commonly isolated from polymicrobial wound infections (53% of 204 polymicrobial cultures). Patients with Enterococcus infections were severely injured with a high proportion of lower extremity amputations and genitourinary injuries. Approximately 65% of polymicrobial Enterococcus infections had other ESKAPE organisms isolated. As biofilms have been suggested as a cause of delayed wound healing, wound infections with persistent recovery of bacteria (isolates of same organism collected ≥14 days apart) and nonrecurrent bacterial isolates were assessed. Biofilm production was significantly associated with recurrent bacteria isolation (97% vs. 59% with nonrecurrent isolates; P < 0.001); however, further analysis is needed to confirm biofilm formation as a predictor of persistent wound infections. CONCLUSIONS The TIDOS MDR/VO Initiative provides comprehensive and detailed data of major microbial threats associated with combat-related wound infections to further the understanding of wound etiology and potentially identify infectious disease countermeasures, which may lead to improvements in combat casualty care.
Collapse
Affiliation(s)
- Katrin Mende
- Infectious Disease Clinical Research Program, Preventive Medicine & Biostatistics Department, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA.,Brooke Army Medical Center, JBSA Fort Sam Houston, TX 78234, USA
| | - Kevin S Akers
- U.S. Army Institute of Surgical Research, JBSA Fort Sam Houston, TX 78234, USA
| | - Stuart D Tyner
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Jason W Bennett
- Multidrug-Resistant Organisms Repository and Surveillance Network, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Mark P Simons
- Naval Medical Research Center, Silver Spring, MD 20910, USA
| | - Dana M Blyth
- Brooke Army Medical Center, JBSA Fort Sam Houston, TX 78234, USA
| | - Ping Li
- Infectious Disease Clinical Research Program, Preventive Medicine & Biostatistics Department, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Laveta Stewart
- Infectious Disease Clinical Research Program, Preventive Medicine & Biostatistics Department, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - David R Tribble
- Infectious Disease Clinical Research Program, Preventive Medicine & Biostatistics Department, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
34
|
Mehdizadeh M, Sheikhpour M, Salahshourifar I, Siadat SD, Saffarian P. An in Vitro Study of Molecular Effects of a Combination Treatment with Antibiotics and Nanofluid Containing Carbon Nano-tubes on Klebsiella pneumoniae. IRANIAN JOURNAL OF PUBLIC HEALTH 2022; 50:2292-2301. [PMID: 35223604 PMCID: PMC8826340 DOI: 10.18502/ijph.v50i11.7585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/09/2021] [Indexed: 11/29/2022]
Abstract
Background: We aimed to prepare a nanofluid, containing f-MWCNTs, and investigate the antibacterial efficacy of f-MWCNTs+ ciprofloxacin (cip) on Klebsiella pneumoniae by evaluating the virulence gene expression. Methods: This study was carried out from 2019 to 2020, in the Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran. The nanofluid containing antibiotic and f-MWCNTs were prepared by the ultrasonic method. The minimum inhibitory concentrations (MICs) of ciprofloxacin and f-MWCNTs were determined using the broth micro dilution MIC tests. For examining the antibacterial effects, the expression level of virulence genes, under the influence of f-MWCNTs, was evaluated by a real-time PCR. Results: The effect of 8 μg/ml ciprofloxacin + 400 μg/ml f-MWCNTs, completely inhibited the growth of the resistant isolate of K. pneumoniae, while, in the ATCC 700,603 isolate, 2 μg/ml ciprofloxacin with 100 μg/ml f-MWCNT could inhibit a bacterial growth. In the resistant K. pneumoniae clinical isolate, after f-MWCNT+cip treatment, the expression of fimA, fimD, wza, and wzi genes was significantly downregulated, compared to the ciprofloxacin treatment, and upregulated, compared to the negative control. For the ATCC 700,603 isolate treated with f-MWCNT+cip, the expression of fimA, fimD and wza virulence genes showed upregulation, compared to the negative control and downregulated in comparison with the ciprofloxacin treatment. Conclusion: Simultaneous treatment of resistant isolate of K. pneumoniae with f-MWCNTs +antibiotic could improve the effectiveness of antibiotic at lower doses, due to the reduced expression of virulence genes in comparison with antibiotic treatment, besides the increased cell wall permeability to antibiotics.
Collapse
Affiliation(s)
- Maryam Mehdizadeh
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mojgan Sheikhpour
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Iman Salahshourifar
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Parvaneh Saffarian
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
35
|
de Jong H, Wösten MMSM, Wennekes T. Sweet impersonators: Molecular mimicry of host glycans by bacteria. Glycobiology 2022; 32:11-22. [PMID: 34939094 PMCID: PMC8881735 DOI: 10.1093/glycob/cwab104] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/03/2021] [Accepted: 09/28/2021] [Indexed: 12/02/2022] Open
Abstract
All bacteria display surface-exposed glycans that can play an important role in their interaction with the host and in select cases mimic the glycans found on host cells, an event called molecular or glycan mimicry. In this review, we highlight the key bacteria that display human glycan mimicry and provide an overview of the involved glycan structures. We also discuss the general trends and outstanding questions associated with human glycan mimicry by bacteria. Finally, we provide an overview of several techniques that have emerged from the discipline of chemical glycobiology, which can aid in the study of the composition, variability, interaction and functional role of these mimicking glycans.
Collapse
Affiliation(s)
- Hanna de Jong
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, and Bijvoet Center for Biomedical Research, Utrecht University, Universiteitsweg 99, Utrecht 3584 CG, The Netherlands
- Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 1, Utrecht 3584 CL, The Netherlands
| | - Marc M S M Wösten
- Department of Biomolecular Health Sciences, Utrecht University, Yalelaan 1, Utrecht 3584 CL, The Netherlands
| | - Tom Wennekes
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, and Bijvoet Center for Biomedical Research, Utrecht University, Universiteitsweg 99, Utrecht 3584 CG, The Netherlands
| |
Collapse
|
36
|
Caseinolytic Proteins (Clp) in the Genus Klebsiella: Special Focus on ClpK. Molecules 2021; 27:molecules27010200. [PMID: 35011428 PMCID: PMC8746953 DOI: 10.3390/molecules27010200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/15/2021] [Accepted: 12/15/2021] [Indexed: 11/16/2022] Open
Abstract
Caseinolytic proteins (Clp), which are present in both prokaryotes and eukaryotes, play a major role in cell protein quality control and survival of bacteria in harsh environmental conditions. Recently, a member of this protein family, ClpK was identified in a pathogenic strain of Klebsiella pneumoniae which was responsible for nosocomial infections. ClpK is linked to the thermal stress survival of this pathogen. The genome wide analysis of Clp proteins in Klebsiella spp. indicates that ClpK is present in only 34% of the investigated strains. This suggests that the uptake of the clpk gene is selective and may only be taken up by a pathogen that needs to survive harsh environmental conditions. In silico analyses and molecular dynamic simulations show that ClpK is mainly α-helical and is highly dynamic. ClpK was successfully expressed and purified to homogeneity using affinity and anion exchange chromatography. Biophysical characterization of ClpK showed that it is predominantly alpha-helical, and this is in agreement with in silico analysis of the protein structure. Furthermore, the purified protein is biologically active and hydrolyses ATP in a concentration- dependent manner.
Collapse
|
37
|
Doorduijn DJ, Heesterbeek DAC, Ruyken M, de Haas CJC, Stapels DAC, Aerts PC, Rooijakkers SHM, Bardoel BW. Polymerization of C9 enhances bacterial cell envelope damage and killing by membrane attack complex pores. PLoS Pathog 2021; 17:e1010051. [PMID: 34752492 PMCID: PMC8604303 DOI: 10.1371/journal.ppat.1010051] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/19/2021] [Accepted: 10/20/2021] [Indexed: 11/21/2022] Open
Abstract
Complement proteins can form membrane attack complex (MAC) pores that directly kill Gram-negative bacteria. MAC pores assemble by stepwise binding of C5b, C6, C7, C8 and finally C9, which can polymerize into a transmembrane ring of up to 18 C9 monomers. It is still unclear if the assembly of a polymeric-C9 ring is necessary to sufficiently damage the bacterial cell envelope to kill bacteria. In this paper, polymerization of C9 was prevented without affecting binding of C9 to C5b-8, by locking the first transmembrane helix domain of C9. Using this system, we show that polymerization of C9 strongly enhanced damage to both the bacterial outer and inner membrane, resulting in more rapid killing of several Escherichia coli and Klebsiella strains in serum. By comparing binding of wildtype and ‘locked’ C9 by flow cytometry, we also show that polymerization of C9 is impaired when the amount of available C9 per C5b-8 is limited. This suggests that an excess of C9 is required to efficiently form polymeric-C9. Finally, we show that polymerization of C9 was impaired on complement-resistant E. coli strains that survive killing by MAC pores. This suggests that these bacteria can specifically block polymerization of C9. All tested complement-resistant E. coli expressed LPS O-antigen (O-Ag), compared to only one out of four complement-sensitive E. coli. By restoring O-Ag expression in an O-Ag negative strain, we show that the O-Ag impairs polymerization of C9 and results in complement-resistance. Altogether, these insights are important to understand how MAC pores kill bacteria and how bacterial pathogens can resist MAC-dependent killing. In this paper, we focus on how complement proteins, an essential part of the immune system, kill Gram-negative bacteria via so-called membrane attack complex (MAC) pores. The MAC is a large pore that consists of five different proteins. The final component, C9, assembles a ring of up to 18 C9 molecules that damages the bacterial cell envelope. Here, we aimed to better understand if this polymeric-C9 ring is necessary to kill bacteria and if bacteria can interfere in its assembly. We uncover that polymerization of C9 increased the damage to the entire bacterial cell envelope, which resulted in more rapid killing of several Gram-negative species. We also show that some clinical Escherichia coli strains can block polymerization of C9 and survive MAC-dependent killing by modifying sugars in the bacterial cell envelope, namely the O-antigen of lipopolysaccharide. These insights help us to better understand how the immune system kills bacteria and how pathogenic bacteria can survive killing.
Collapse
Affiliation(s)
- Dennis J. Doorduijn
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dani A. C. Heesterbeek
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maartje Ruyken
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Carla J. C. de Haas
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Daphne A. C. Stapels
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Piet C. Aerts
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Suzan H. M. Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bart W. Bardoel
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
38
|
Baghbanijavid S, Kafil HS, Farajniya S, Moaddab SR, Hosainzadegan H, Sefidan FY, Varshouchi M, Leylabadlo HE, Ghotaslou R. The Association of the Phylogenetic Typing of the Klebsiella pneumoniae Isolates with Antibiotic Resistance. Emerg Med Int 2021; 2021:1316992. [PMID: 34777863 PMCID: PMC8589475 DOI: 10.1155/2021/1316992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/09/2021] [Accepted: 10/22/2021] [Indexed: 11/17/2022] Open
Abstract
Klebsiella pneumoniae complex (KPC) accounts for approximately one-third of all Gram-negative infections. Moreover, it is highly resistant and can taxonomically be distributed into KpI, KpII, and KpIII phylogroups. This study aimed to investigate the distribution of phylogenetic groups and the relationship between them and antibiotic resistance patterns. For this purpose, we collected KPC isolates from Tabriz, Iran, between 2018 and 2020. Antimicrobial susceptibility testing was performed by disk diffusion agar, and phylogenetic groups were then examined using gyrA restriction fragment length polymorphism (RFLP) and parC PCR methods. A total of 100 KPC isolates were obtained from the clinical specimens (urine, respiratory secretion, blood, wounds, and trachea). The enrolled patients included 47 men and 53 women aged from 1 to 91 years old. The highest sensitivity was found related to fosfomycin as 85%, followed by amikacin as 66%. The three phylogenetically groups by the RFLP-PCR method were found in KPC, 96% (96 isolates) as KpI, 3% (3 isolates) as KpII, and 1% (1isolate) as KpIII. The highest antibiotic resistance was observed in KpI. It was shown that a valid identification of three phylogenetic groups of KPC can be done by combining both gyrA PCR-RFLP and parC PCR. Of note, the KpI group was also observed as the dominant phylogenetic group with the highest resistance to antibiotics.
Collapse
Affiliation(s)
- Shabnam Baghbanijavid
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Samadi Kafil
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safar Farajniya
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Reza Moaddab
- Department of Laboratory Sciences, Faculty of Paramedicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hasan Hosainzadegan
- Department of Microbiology, Faculty of Medicine, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Fatemeh Yeganeh Sefidan
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mojtaba Varshouchi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Reza Ghotaslou
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
39
|
In vitro activity of carvacrol in combination with meropenem against carbapenem-resistant Klebsiella pneumoniae. Folia Microbiol (Praha) 2021; 67:143-156. [PMID: 34729712 DOI: 10.1007/s12223-021-00908-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 08/04/2021] [Indexed: 10/19/2022]
Abstract
Carbapenem resistance observed in Klebsiella pneumoniae strains limits treatment options. Therefore, use of antibiotics combined with bioactive compounds may be an important strategy to control K. pneumoniae. The purpose of this study was to evaluate the activity of combination of carvacrol and meropenem on carbapenem-resistant K. pneumoniae (CRKP) strains. The presence of blaOXA-48 carbapenemase in all 25 CRKP strains was identified using the PCR technique. The combination of carvacrol and meropenem was tested for antimicrobial activity on CRKP strains. The minimum inhibitory concentrations of carvacrol and meropenem were detected within a range of 32-128 µg/mL using the broth microdilution method. Synergy between carvacrol and meropenem was observed on 8 of the 25 CRKP strains by checkerboard assay (FICI = 0.5) and confirmed by time-kill assay. According to the live-dead test results, the viability percentage of the cells exposed to synergistic combination was 35.47% at the end of 24 h. The membrane damage caused by the synergistic combination was spectrophotometrically measured (A = 0.21) and further confirmed by SEM analysis. According to the MTT assay, both carvacrol and meropenem did not show any statistically significant cytotoxic effect on Vero cells (p > 0.05). In conclusion, the results suggest that carvacrol and meropenem can act synergistically to inhibit the growth of CRKP.
Collapse
|
40
|
Kaszowska M, Majkowska-Skrobek G, Markwitz P, Lood C, Jachymek W, Maciejewska A, Lukasiewicz J, Drulis-Kawa Z. The Mutation in wbaP cps Gene Cluster Selected by Phage-Borne Depolymerase Abolishes Capsule Production and Diminishes the Virulence of Klebsiella pneumoniae. Int J Mol Sci 2021; 22:11562. [PMID: 34768992 PMCID: PMC8583740 DOI: 10.3390/ijms222111562] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 12/20/2022] Open
Abstract
Klebsiella pneumoniae is considered one of the most critical multidrug-resistant pathogens and urgently requires new therapeutic strategies. Capsular polysaccharides (CPS), lipopolysaccharides (LPS), and exopolysaccharides (EPS) are the major virulence factors protecting K. pneumoniae against the immune response and thus may be targeted by phage-based therapeutics such as polysaccharides-degrading enzymes. Since the emergence of resistance to antibacterials is generally considered undesirable, in this study, the genetic and phenotypic characteristics of resistance to the phage-borne CPS-degrading depolymerase and its effect on K. pneumoniae virulence were investigated. The K63 serotype targeting depolymerase (KP36gp50) derived from Klebsiella siphovirus KP36 was used as the selective agent during the treatment of K. pneumoniae 486 biofilm. Genome-driven examination combined with the surface polysaccharide structural analysis of resistant mutant showed the point mutation and frameshift in the wbaP gene located within the cps gene cluster, resulting in the loss of the capsule. The sharp decline in the yield of CPS was accompanied by the production of a larger amount of smooth LPS. The modification of the surface polysaccharide layers did not affect bacterial fitness nor the insensitivity to serum complement; however, it made bacteria more prone to phagocytosis combined with the higher adherence and internalization to human lung epithelial cells. In that context, it was showed that the emerging resistance to the antivirulence agent (phage-borne capsule depolymerase) results in beneficial consequences, i.e., the sensitization to the innate immune response.
Collapse
Affiliation(s)
- Marta Kaszowska
- Laboratory of Microbial Immunochemistry and Vaccines, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.K.); (W.J.); (A.M.)
| | - Grazyna Majkowska-Skrobek
- Department of Pathogen Biology and Immunology, University of Wroclaw, 51-148 Wroclaw, Poland; (G.M.-S.); (P.M.)
| | - Pawel Markwitz
- Department of Pathogen Biology and Immunology, University of Wroclaw, 51-148 Wroclaw, Poland; (G.M.-S.); (P.M.)
| | - Cédric Lood
- Department of Microbial and Molecular Systems, KU Leuven, 3001 Leuven, Belgium;
- Department of Biosystems, KU Leuven, 3001 Leuven, Belgium
| | - Wojciech Jachymek
- Laboratory of Microbial Immunochemistry and Vaccines, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.K.); (W.J.); (A.M.)
| | - Anna Maciejewska
- Laboratory of Microbial Immunochemistry and Vaccines, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.K.); (W.J.); (A.M.)
| | - Jolanta Lukasiewicz
- Laboratory of Microbial Immunochemistry and Vaccines, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (M.K.); (W.J.); (A.M.)
| | - Zuzanna Drulis-Kawa
- Department of Pathogen Biology and Immunology, University of Wroclaw, 51-148 Wroclaw, Poland; (G.M.-S.); (P.M.)
| |
Collapse
|
41
|
Zhang W, Feng Y, Zhao H, Yan C, Feng J, Gan L, Cui J, Liu S, Zhang R, Du S, Li N, Xu W, Han J, Li R, Xue G, Yuan J. A Recombinase Aided Amplification Assay for Rapid Detection of the Klebsiella pneumoniae Carbapenemase Gene and Its Characteristics in Klebsiella pneumoniae. Front Cell Infect Microbiol 2021; 11:746325. [PMID: 34616692 PMCID: PMC8488121 DOI: 10.3389/fcimb.2021.746325] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/24/2021] [Indexed: 11/20/2022] Open
Abstract
Klebsiella pneumoniae carbapenemase genes (blaKPC) play an important role in carbapenem-resistant Enterobacteriaceae in China. A rapid detection method for blaKPC genes and investigations into the molecular characteristics of blaKPC positive Klebsiella pneumoniae were necessary. In this study, an easy and rapid recombinase aided amplification assay (RAA) for blaKPC was established. This protocol could be completed at 39°C in 15–20 min. The sensitivity of this assay was determined as 48 copies per reaction, and the specificity was 100%. The blaKPC RAA method could be used for clinical diagnosis and epidemiological investigation. Among 801 fecal samples from inpatients, 34 blaKPC positive isolates were identified from each sample, of which 23 isolates were K. pneumoniae. ST11 with blaKPC-2 was the most prevalent type. All these strains were multidrug resistant and carried various virulence genes. Fecal carriage of blaKPC positive carbapenem-resistant K.pneumoniae poses significant challenges for public health control.
Collapse
Affiliation(s)
- Weiwei Zhang
- The Second Affiliated Hospital of Dalian Medical University, Liaoning, China.,Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Yanling Feng
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Hanqing Zhao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Chao Yan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Junxia Feng
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Lin Gan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Jinghua Cui
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Shiyu Liu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Rui Zhang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Shuheng Du
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Nannan Li
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Wenjian Xu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Juqiang Han
- Department of Daily Clinic, Seventh Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Rongkuan Li
- The Second Affiliated Hospital of Dalian Medical University, Liaoning, China
| | - Guanhua Xue
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| | - Jing Yuan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
42
|
Ali S, Alam M, Hasan GM, Hassan MI. Potential therapeutic targets of Klebsiella pneumoniae: a multi-omics review perspective. Brief Funct Genomics 2021; 21:63-77. [PMID: 34448478 DOI: 10.1093/bfgp/elab038] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 11/15/2022] Open
Abstract
The multidrug resistance developed in many organisms due to the prolonged use of antibiotics has been an increasing global health crisis. Klebsiella pneumoniae is a causal organism for various infections, including respiratory, urinary tract and biliary diseases. Initially, immunocompromised individuals are primarily affected by K. pneumoniae. Due to the emergence of hypervirulent strains recently, both healthy and immunocompetent individuals are equally susceptible to K. pneumoniae infections. The infections caused by multidrug-resistant and hypervirulent K. pneumoniae strains are complicated to treat, illustrating an urgent need to develop novel and more practical approaches to combat the pathogen. We focused on the previously performed high-throughput analyses by other groups to discover several novel enzymes that may be considered attractive drug targets of K. pneumoniae. These targets qualify most of the selection criteria for drug targeting, including an absence of its homolog's gene in the host. The capsule, lipopolysaccharide, fimbriae, siderophores and essential virulence factors facilitate the pathogen entry, infection and survival inside the host. This review discusses K. pneumoniae pathophysiology, including its virulence determinants and further the potential drug targets that might facilitate the discovery of novel drugs and effective treatment regimens shortly.
Collapse
Affiliation(s)
- Sabeeha Ali
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar New Delhi 110025, India
| | - Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar New Delhi 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar New Delhi 110025, India
| |
Collapse
|
43
|
Walker SS, Black TA. Are outer-membrane targets the solution for MDR Gram-negative bacteria? Drug Discov Today 2021; 26:2152-2158. [PMID: 33798647 DOI: 10.1016/j.drudis.2021.03.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/27/2021] [Accepted: 03/23/2021] [Indexed: 01/17/2023]
Abstract
The outer membrane (OM) of Gram-negative bacteria confers a significant barrier to many antibacterial agents targeting periplasmic and cytosolic functions. 'Synergist' approaches to disrupt the OM have been hampered by poor specificity and accompanying toxicities. The OM contains proteins required for optimal growth and pathogenesis, including lipopolysaccharide (LPS) and capsular polysaccharide (CPS) transport, porins for uptake of macromolecules, and transporters for essential elements (such as iron). Does the external proximity of these proteins offer an enhanced potential to identify effective therapies? Here, we review recent experiences in exploiting Gram-negative OM proteins (OMPs) to address the calamity of exploding antimicrobial resistance. Teaser: Multidrug-resistant (MDR) Gram-negative bacteria are a growing crisis. Few new antimicrobial chemotypes or targets have been identified after decades of screening. Are OMP targets a solution to MDR Gram-negative bacteria?
Collapse
Affiliation(s)
- Scott S Walker
- Infectious Diseases and Vaccines Basic Research, Merck & Co., Inc, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Todd A Black
- Infectious Diseases and Vaccines Basic Research, Merck & Co., Inc, 770 Sumneytown Pike, West Point, PA 19486, USA.
| |
Collapse
|
44
|
Majkowska-Skrobek G, Markwitz P, Sosnowska E, Lood C, Lavigne R, Drulis-Kawa Z. The evolutionary trade-offs in phage-resistant Klebsiella pneumoniae entail cross-phage sensitization and loss of multidrug resistance. Environ Microbiol 2021; 23:7723-7740. [PMID: 33754440 DOI: 10.1111/1462-2920.15476] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/11/2021] [Accepted: 03/18/2021] [Indexed: 12/18/2022]
Abstract
Bacteriophage therapy is currently being evaluated as a critical complement to traditional antibiotic treatment. However, the emergence of phage resistance is perceived as a major hurdle to the sustainable implementation of this antimicrobial strategy. By combining comprehensive genomics and microbiological assessment, we show that the receptor-modification resistance to capsule-targeting phages involves either escape mutation(s) in the capsule biosynthesis cluster or qualitative changes in exopolysaccharides, converting clones to mucoid variants. These variants introduce cross-resistance to phages specific to the same receptor yet sensitize to phages utilizing alternative ones. The loss/modification of capsule, the main Klebsiella pneumoniae virulence factor, did not dramatically impact population fitness, nor the ability to protect bacteria against the innate immune response. Nevertheless, the introduction of phage drives bacteria to expel multidrug resistance clusters, as observed by the large deletion in K. pneumoniae 77 plasmid containing blaCTX-M , ant(3″), sul2, folA, mph(E)/mph(G) genes. The emerging bacterial resistance to viral infection steers evolution towards desired population attributes and highlights the synergistic potential for combined antibiotic-phage therapy against K. pneumoniae.
Collapse
Affiliation(s)
- Grazyna Majkowska-Skrobek
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, Wroclaw, Poland
| | - Pawel Markwitz
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, Wroclaw, Poland
| | - Ewelina Sosnowska
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, Wroclaw, Poland
| | - Cédric Lood
- Department of Biosystems, Laboratory of Gene Technology, KU Leuven, 3001 Heverlee, Belgium.,Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics, Laboratory of Computational Systems Biology, KU Leuven, 3000 Leuven, Belgium
| | - Rob Lavigne
- Department of Biosystems, Laboratory of Gene Technology, KU Leuven, 3001 Heverlee, Belgium
| | - Zuzanna Drulis-Kawa
- Department of Pathogen Biology and Immunology, Institute of Genetics and Microbiology, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
45
|
Hower S, McCormack R, Bartra SS, Alonso P, Podack ER, Shembade N, Plano GV. LPS modifications and AvrA activity of Salmonella enterica serovar Typhimurium are required to prevent Perforin-2 expression by infected fibroblasts and intestinal epithelial cells. Microb Pathog 2021; 154:104852. [PMID: 33762201 DOI: 10.1016/j.micpath.2021.104852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 10/21/2022]
Abstract
Cellular Perforin-2 (MPEG1) is a pore-forming MACPF family protein that plays a critical role in the defense against bacterial pathogens. Macrophages, neutrophils, and several other cell types that are part of the front line of innate defenses constitutively express high levels of Perforin-2; whereas, most other cell types must be induced to express Perforin-2 by interferons (α, β and γ) and/or PAMPs such as LPS. In this study, we demonstrate that many bacterial pathogens can limit the expression of Perforin-2 in cells normally inducible for Perforin-2 expression, while ordinarily commensal or non-pathogenic bacteria triggered high levels of Perforin-2 expression in these same cell types. The mechanisms by which pathogens suppress Perforin-2 expression was explored further using Salmonella enterica serovar Typhimurium and cultured MEFs as well as intestinal epithelial cell lines. These studies identified multiple factors required to minimize the expression of Perforin-2 in cell types inducible for Perforin-2 expression. These included the PmrAB and PhoPQ two-component systems, select LPS modification enzymes and the Type III secretion effector protein AvrA.
Collapse
Affiliation(s)
- Suzanne Hower
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Ryan McCormack
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Sara Schesser Bartra
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Patricia Alonso
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Eckhard R Podack
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, 33136, USA
| | - Noula Shembade
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, 33136, USA
| | - Gregory V Plano
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
46
|
Abstract
Klebsiella pneumoniae are Gram-negative facultative anaerobes that are found within host-associated commensal microbiomes, but they can also cause a wide range of infections that are often difficult to treat. These infections are caused by different pathotypes of K. pneumoniae, called either classical or hypervirulent strains. Klebsiella pneumoniae are Gram-negative facultative anaerobes that are found within host-associated commensal microbiomes, but they can also cause a wide range of infections that are often difficult to treat. These infections are caused by different pathotypes of K. pneumoniae, called either classical or hypervirulent strains. These two groups are genetically distinct, inhabit nonoverlapping geographies, and cause different types of harmful infections in humans. These distinct bacterial groups have also been found to interact differently with the host immune system. Initial innate immune defenses against K. pneumoniae infection include complement, macrophages, neutrophils, and monocytes; these defenses are primary strategies employed by the host to clear infections. K. pneumoniae pathogenesis depends upon the interactions between the microbe and each of these host defenses, and it is becoming increasingly apparent that bacterial genetic diversity impacts the outcomes of these interactions. Here, we highlight recent advances in our understanding of K. pneumoniae pathogenesis, with a focus on how bacterial evolution and diversity impact K. pneumoniae interactions with mammalian innate immune host defenses. We also discuss outstanding questions regarding how K. pneumoniae can frustrate normal immune responses, capitalize upon states of immunocompromise, and cause infections with high mortality.
Collapse
|
47
|
Buffet A, Rocha EPC, Rendueles O. Nutrient conditions are primary drivers of bacterial capsule maintenance in Klebsiella. Proc Biol Sci 2021; 288:20202876. [PMID: 33653142 PMCID: PMC7935059 DOI: 10.1098/rspb.2020.2876] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 02/05/2021] [Indexed: 02/06/2023] Open
Abstract
The fitness cost associated with the production of bacterial capsules is considered to be offset by the protection provided by these extracellular structures against biotic aggressions or abiotic stress. However, it is unknown if the capsule contributes to fitness in the absence of these. Here, we explored conditions favouring the maintenance of the capsule in Klebsiella pneumoniae, where the capsule is known to be a major virulence factor. Using short-term experimental evolution on different Klebsiella strains, we showed that small environmental variations have a strong impact on the maintenance of the capsule. Capsule inactivation is frequent in nutrient-rich, but scarce in nutrient-poor media. Competitions between wild-type and capsule mutants in nine different strains confirmed that the capsule is costly in nutrient-rich media. Surprisingly, these results also showed that the presence of a capsule provides a clear fitness advantage in nutrient-poor conditions by increasing both growth rates and population yields. The comparative analyses of the wild-type and capsule mutants reveal complex interactions between the environment, genetic background and serotype even in relation to traits known to be relevant during pathogenesis. In conclusion, our data suggest there are novel roles for bacterial capsules yet to be discovered and further supports the notion that the capsule's role in virulence may be a by-product of its contribution to bacterial adaptation outside the host.
Collapse
Affiliation(s)
- Amandine Buffet
- Microbial Evolutionary Genomics, Institut Pasteur, CNRS, UMR3525, Paris 75015, France
| | - Eduardo P. C. Rocha
- Microbial Evolutionary Genomics, Institut Pasteur, CNRS, UMR3525, Paris 75015, France
| | - Olaya Rendueles
- Microbial Evolutionary Genomics, Institut Pasteur, CNRS, UMR3525, Paris 75015, France
| |
Collapse
|
48
|
Tummillo KM, Hazlett KR. Co-Opting Host Receptors for Targeted Delivery of Bioconjugates-From Drugs to Bugs. Molecules 2021; 26:molecules26051479. [PMID: 33803208 PMCID: PMC7963163 DOI: 10.3390/molecules26051479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 02/26/2021] [Accepted: 03/04/2021] [Indexed: 11/16/2022] Open
Abstract
Bioconjugation has allowed scientists to combine multiple functional elements into one biological or biochemical unit. This assembly can result in the production of constructs that are targeted to a specific site or cell type in order to enhance the response to, or activity of, the conjugated moiety. In the case of cancer treatments, selectively targeting chemotherapies to the cells of interest limit harmful side effects and enhance efficacy. Targeting through conjugation is also advantageous in delivering treatments to difficult-to-reach tissues, such as the brain or infections deep in the lung. Bacterial infections can be more selectively treated by conjugating antibiotics to microbe-specific entities; helping to avoid antibiotic resistance across commensal bacterial species. In the case of vaccine development, conjugation is used to enhance efficacy without compromising safety. In this work, we will review the previously mentioned areas in which bioconjugation has created new possibilities and advanced treatments.
Collapse
Affiliation(s)
- Kristen M. Tummillo
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA;
- Admera Health, South Plainfield, NJ 07080, USA
| | - Karsten R.O. Hazlett
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208, USA;
- Correspondence: ; Tel.: +1-518-262-2338
| |
Collapse
|
49
|
Mike LA, Stark AJ, Forsyth VS, Vornhagen J, Smith SN, Bachman MA, Mobley HLT. A systematic analysis of hypermucoviscosity and capsule reveals distinct and overlapping genes that impact Klebsiella pneumoniae fitness. PLoS Pathog 2021; 17:e1009376. [PMID: 33720976 PMCID: PMC7993769 DOI: 10.1371/journal.ppat.1009376] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/25/2021] [Accepted: 02/12/2021] [Indexed: 12/11/2022] Open
Abstract
Hypervirulent K. pneumoniae (hvKp) is a distinct pathotype that causes invasive community-acquired infections in healthy individuals. Hypermucoviscosity (hmv) is a major phenotype associated with hvKp characterized by copious capsule production and poor sedimentation. Dissecting the individual functions of CPS production and hmv in hvKp has been hindered by the conflation of these two properties. Although hmv requires capsular polysaccharide (CPS) biosynthesis, other cellular factors may also be required and some fitness phenotypes ascribed to CPS may be distinctly attributed to hmv. To address this challenge, we systematically identified genes that impact capsule and hmv. We generated a condensed, ordered transposon library in hypervirulent strain KPPR1, then evaluated the CPS production and hmv phenotypes of the 3,733 transposon mutants, representing 72% of all open reading frames in the genome. We employed forward and reverse genetic screens to evaluate effects of novel and known genes on CPS biosynthesis and hmv. These screens expand our understanding of core genes that coordinate CPS biosynthesis and hmv, as well as identify central metabolism genes that distinctly impact CPS biosynthesis or hmv, specifically those related to purine metabolism, pyruvate metabolism and the TCA cycle. Six representative mutants, with varying effect on CPS biosynthesis and hmv, were evaluated for their impact on CPS thickness, serum resistance, host cell association, and fitness in a murine model of disseminating pneumonia. Altogether, these data demonstrate that hmv requires both CPS biosynthesis and other cellular factors, and that hmv and CPS may serve distinct functions during pathogenesis. The integration of hmv and CPS to the metabolic status of the cell suggests that hvKp may require certain nutrients to specifically cause deep tissue infections.
Collapse
Affiliation(s)
- Laura A. Mike
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Andrew J. Stark
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Valerie S. Forsyth
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jay Vornhagen
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Sara N. Smith
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Michael A. Bachman
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Harry L. T. Mobley
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
50
|
Miran W, Naradasu D, Okamoto A. Pathogens electrogenicity as a tool for in-situ metabolic activity monitoring and drug assessment in biofilms. iScience 2021; 24:102068. [PMID: 33554070 PMCID: PMC7859304 DOI: 10.1016/j.isci.2021.102068] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Concerns regarding increased antibiotic resistance arising from the emergent properties of biofilms have spurred interest in the discovery of novel antibiotic agents and techniques to directly estimate metabolic activity in biofilms. Although a number of methods have been developed to quantify biofilm formation, real-time quantitative assessment of metabolic activity in label-free biofilms remains a challenge. Production of electrical current via extracellular electron transport (EET) has recently been found in pathogens and appears to correlate with their metabolic activity. Accordingly, monitoring the production of electrical currents as an indicator of cellular metabolic activity in biofilms represents a new direction for research aiming to assess and screen the effects of antimicrobials on biofilm activity. In this article, we reviewed EET-capable pathogens and the methods to monitor biofilm activity to discuss advantages of using the capability of pathogens to produce electrical currents and effective combination of these methods. Moreover, we discussed EET mechanisms by pathogenic and environmental bacteria and open questions for the physiological roles of EET in pathogen's biofilm. The present limitations and possible future directions of in situ biofilm metabolic activity assessment for large-scale screening of antimicrobials are also discussed.
Collapse
Affiliation(s)
- Waheed Miran
- International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Divya Naradasu
- International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Akihiro Okamoto
- International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, North 13 West 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
| |
Collapse
|