1
|
García-González N, Beamud B, Sevilla-Fortuny J, Sánchez-Hellín V, Vidal I, Rodríguez JC, Fuster B, Tormo N, Salvador C, Gimeno C, Gomila-Sard B, Giner S, Martínez O, Colomina J, Navarro D, Domínguez V, González-Candelas F. Genomic surveillance reveals different transmission patterns between third-generation cephalosporin and carbapenem resistance in Klebsiella pneumoniae in the Comunidad Valenciana (Spain), 2018-2020. Antimicrob Resist Infect Control 2025; 14:44. [PMID: 40336117 PMCID: PMC12060429 DOI: 10.1186/s13756-025-01553-2] [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: 11/08/2024] [Accepted: 04/08/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND The emergence and spread of third-generation cephalosporins (3GC) and carbapenem-resistant Klebsiella pneumoniae pose a global critical challenge. Understanding the transmission dynamics within and between hospital environments is crucial to develop effective control strategies. METHODS From 2017 to 2019, we conducted a genomic surveillance program in eight hospitals of the Comunitat Valenciana, Spain, collecting and sequencing 1,768 3GC- and carbapenem-resistant isolates. We quantified the overall transmission using core genomes and assessed the contribution of national and global isolates to the spread of AMR in the region by including 11,967 database genomes in the analysis. RESULTS The local collection was highly diverse, involving 188 lineages, including global high-risk clones such as ST307 and ST11, and 3GC and carbapenem resistance determinants. Half of the isolates were involved in transmission, with 70.5% occurring within hospitals. CONCLUSIONS Different transmission patterns characterized the spread of 3GC- and carbapenem resistance in the region. While inter-hospital transmission played a significant role in the spread of 3GC-resistance, this was only sporadic for carbapenem resistance. Moreover, the factors behind inter-hospital spread for each type of resistance differed: while 3GC-resistance likely disseminated between hospitals through intermediate steps, carbapenem resistance was driven by more direct transmission routes. The burden of national and global cases on the ongoing regional AMR dissemination was low. Moreover, we revealed the rapid expansion in the region and globally of lineage ST307 carrying the blaCTX-M-15 gene, a main driver of local transmissions, providing a deeper understanding of the successful spread of this high-risk clone.
Collapse
Affiliation(s)
- Neris García-González
- Unidad Mixta Infección y Salud Pública FISABIO-Univ. València, Instituto Biología Integrativa de Sistemas (CSIC-UV), Valencia, Spain.
| | - Beatriz Beamud
- Unidad Mixta Infección y Salud Pública FISABIO-Univ. València, Instituto Biología Integrativa de Sistemas (CSIC-UV), Valencia, Spain
- Synthetic Biology Unit, Institut Pasteur, Paris, France
| | - Jordi Sevilla-Fortuny
- Unidad Mixta Infección y Salud Pública FISABIO-Univ. València, Instituto Biología Integrativa de Sistemas (CSIC-UV), Valencia, Spain
| | | | | | | | - Begoña Fuster
- Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Nuria Tormo
- Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Carme Salvador
- Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Concepción Gimeno
- Consorcio Hospital General Universitario de Valencia, Valencia, Spain
- Dpto. Microbiología y Ecología, Universitat de València, Valencia, Spain
| | | | - Salvador Giner
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | | | - David Navarro
- Dpto. Microbiología y Ecología, Universitat de València, Valencia, Spain
- Hospital Clínico Universitario de Valencia, Valencia, Spain
- CIBER en Enfermedades Infecciosas, Madrid, Spain
| | | | - Fernando González-Candelas
- Unidad Mixta Infección y Salud Pública FISABIO-Univ. València, Instituto Biología Integrativa de Sistemas (CSIC-UV), Valencia, Spain.
- CIBER en Epidemiología y Salud Pública, Madrid, Spain.
| |
Collapse
|
2
|
Pu X, Wang X, Wang J, Gu Z, Zhu H, Li C. Microbial and metabolic profiles associated with HPV infection and cervical intraepithelial neoplasia: a multi-omics study. Microbiol Spectr 2025:e0019225. [PMID: 40304477 DOI: 10.1128/spectrum.00192-25] [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: 01/20/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
Cervical cancer is the most common malignancy of the female reproductive system, with the incidence of human papillomavirus (HPV) being a crucial factor in its pathogenesis. Emerging evidence indicates that cervicovaginal microbiota may influence HPV persistence and cervical intraepithelial neoplasia (CIN). However, the interplay between cervicovaginal and cervical tissue microbiomes and their association with HPV infection and CIN remains poorly understood. In this cross-sectional study, we analyzed the microbiota profiles of cervicovaginal and cervical tissue via five-region 16S rRNA gene metabarcoding, along with cervicovaginal metabolites, including short-chain fatty acids (SCFAs) and non-targeted metabolomic data, from 94 women. Key species, particularly Lacticaseibacillus and various anaerobes, are vital components of the microbiota found in both cervicovaginal secretions and cervical tissue, despite notable differences in microbial composition. The CIN group exhibited significant differences in microbial diversity and composition compared to the control groups, with key species such as Lacticaseibacillus iners and Prevotella bivia associated with HPV status and CIN progression. Metabolomic analysis revealed alterations in glycerophospholipid metabolism, but not in SCFAs, with correlations observed between metabolites and HPV status. Notable associations, including P. bivia-PE(18:1/0:0)-HPV and Fusobacterium periodonticum-PI(40:6)-HPV, were identified. Our findings emphasize the critical roles of cervicovaginal and cervical tissue microbiomes in HPV infection and CIN development, highlighting specific microbial species and metabolic pathways for early detection and therapeutic targets. IMPORTANCE Cervical cancer is the most prevalent malignancy in the female reproductive system, with human papillomavirus (HPV) persistency being a critical factor in its pathogenesis. This study highlights the significant yet often overlooked role of cervicovaginal secretion and cervical tissue microbiota in influencing HPV infection and the progression of cervical intraepithelial neoplasia (CIN). By employing a multi-omics approach, we elucidated distinct microbiota profiles in cervical tissues compared to cervicovaginal secretions, revealing a complex interplay between specific bacterial species (notably Lacticaseibacillus and anaerobes) and metabolomic changes associated with glycerophospholipid metabolism. Our findings address a significant gap in understanding the interplay between cervicovaginal secretion and cervical intratissue microbiomes, HPV infection, and CIN.
Collapse
Affiliation(s)
- Xiaowen Pu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiao Wang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jingjing Wang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhengrong Gu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haiyan Zhu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chao Li
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
3
|
Huynh MT, Jusselme MD, Van Pham H, Nguyen TD, Chau MQ. Characterization of phenotypic and genotypic traits of Klebsiella pneumoniae strains resistant to 3rd generation cephalosporins in hospital settings: A case study in Ho Chi Minh City, Vietnam. Mol Biol Rep 2025; 52:396. [PMID: 40232586 PMCID: PMC12000183 DOI: 10.1007/s11033-025-10493-4] [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: 01/10/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Klebsiella pneumoniae is a dangerous pathogen, responsible for a variety of infections and has been growing resistance to antibiotics, making it difficult to treat diseases caused by this bacterium. A better understanding of the phenotypic and genotypic characteristics of this bacterium is essential for developing more effective treatments. This study examines the prevalence of Klebsiella pneumoniae in patient samples from eight hospitals in Ho Chi Minh City, Vietnam, in 2023, focusing on its AmpC β-lactamase, ESBL, and carbapenemase genotypes. METHODS AND RESULTS A total of 230 Klebsiella pneumoniae strains were isolated from clinical specimens, including sputum, pus, urine, bronchial lavage fluid, blood, and other fluids. The disk diffusion method detected the presence of these resistance enzymes, while multiplex real-time PCR identified the associated genes. Results showed that 78.7% of isolates produced AmpC β-lactamase, 21.3% produced ESBL, and 27.0% produced carbapenemase. ESBL-producing strains (90.9%) were more common than AmpC (67.4%) and carbapenemase (45.7%) strains. Additionally, 27.0% of isolates produced multiple resistance enzymes. The most prevalent AmpC gene was DHA (63.5%), while SHV (84.3%), TEM (55.2%), and CTX-M (70.0%) were the most common ESBL genes. OXA-48 (31.3%) was the dominant carbapenemase gene, followed by NDM1 (24.8%) and KPC (9.1%). On average, each strain carried 3.3 resistance genes. CONCLUSIONS This study reveals a high prevalence of antimicrobial resistance in Klebsiella pneumoniae isolates from patient samples in Ho Chi Minh City hospitals, notably to third-generation cephalosporins and carbapenems. These findings highlight the need for ongoing surveillance and improved treatment strategies.
Collapse
Affiliation(s)
- Minh Tuan Huynh
- Medical Microbiology Department, University Medical Center Ho Chi Minh City, 215 Hong Bang street, Ward 11, District 5, Ho Chi Minh City, Vietnam
- Department of Microbiology and Parasitology, University of Medicine and Pharmacy at Ho Chi Minh City, 217 Hong Bang street, Ward 11, District 5, Ho Chi Minh City, Vietnam
| | - My Dung Jusselme
- LEESU, Univ Paris Est Creteil, ENPC, Institut Polytechnique de Paris, 61 avenue du Général de Gaulle, 94000, Creteil, France.
| | - Hung Van Pham
- Nam Khoa Trading and Service Company Limited, 793/58 Tran Xuan Soan street, Tan Hung Ward, District 7, Ho Chi Minh City, Vietnam
| | - Tien Dung Nguyen
- National Fisheries Inspection and Quanlity Assurance Branch 4, 271 To Ngoc Van street, Linh Dong Ward, Thu Duc City, Ho Chi Minh City, Vietnam
| | - Minh Quynh Chau
- Medical Microbiology Department, University Medical Center Ho Chi Minh City, 215 Hong Bang street, Ward 11, District 5, Ho Chi Minh City, Vietnam.
| |
Collapse
|
4
|
Grieshop MP, Behr AA, Bowden S, Lin JD, Molari M, Reynolds GZ, Brooks EF, Doyle B, Rodriguez-Nava G, Salinas JL, Banaei N, Bhatt AS. Replicative selfish genetic elements are driving rapid pathogenic adaptation of Enterococcus faecium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.16.643550. [PMID: 40161577 PMCID: PMC11952509 DOI: 10.1101/2025.03.16.643550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Understanding how healthcare-associated pathogens adapt in clinical environments can inform strategies to reduce their burden. Here, we investigate the hypothesis that insertion sequences (IS), prokaryotic transposable elements, are a dominant mediator of rapid genomic evolution in healthcare-associated pathogens. Among 28,207 publicly available pathogen genomes, we find high copy numbers of the replicative ISL3 family in healthcare-associated Enterococcus faecium, Streptococcus pneumoniae and Staphylococcus aureus. In E. faecium, the ESKAPE pathogen with the highest IS density, we find that ISL3 proliferation has increased in the last 30 years. To enable better identification of structural variants, we long read-sequenced a new, single hospital collection of 282 Enterococcal infection isolates collected over three years. In these samples, we observed extensive, ongoing structural variation of the E. faecium genome, largely mediated by active replicative ISL3 elements. To determine if ISL3 is actively replicating in clinical timescales in its natural, gut microbiome reservoir, we long read-sequenced a collection of 28 longitudinal stool samples from patients undergoing hematopoietic cell transplantation, whose gut microbiomes were dominated by E. faecium. We found up to six structural variants of a given E. faecium strain within a single stool sample. Examining longitudinal samples from one individual in further detail, we find ISL3 elements can replicate and move to specific positions with profound regulatory effects on neighboring gene expression. In particular, we identify an ISL3 element that upon insertion replaces an imperfect -35 promoter sequence at a folT gene locus with a perfect -35 sequence, which leads to substantial upregulation of expression of folT, driving highly effective folate scavenging. As a known folate auxotroph, E. faecium depends on other members of the microbiota or diet to supply folate. Enhanced folate scavenging may enable E. faecium to thrive in the setting of microbiome collapse that is common in HCT and other critically ill patients. Together, ISL3 expansion has enabled E. faecium to rapidly evolve in healthcare settings, and this likely contributes to its metabolic fitness and may strongly influence its ongoing trajectory of genomic evolution.
Collapse
Affiliation(s)
- Matthew P Grieshop
- Department of Genetics, Stanford University, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford, CA, USA
- Stanford University School of Medicine, Stanford, CA, USA
| | - Aaron A Behr
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Sierra Bowden
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Jordan D Lin
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
| | - Marco Molari
- Swiss Institute of Bioinformatics, Basel, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Gabriella Zm Reynolds
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
| | - Erin F Brooks
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
- Current Address: School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Boryana Doyle
- Stanford University School of Medicine, Stanford, CA, USA
| | - Guillermo Rodriguez-Nava
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA
| | - Jorge L Salinas
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA
| | - Niaz Banaei
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA
- Clinical Microbiology Laboratory, Stanford University Medical Center, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Ami S Bhatt
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University, Stanford, CA, USA
| |
Collapse
|
5
|
Aguilar-Bultet L, Gómez-Sanz E, García-Martín AB, Hug MA, Furger R, Eichenberger L, Bagutti C, Tschudin-Sutter S. Extended-Spectrum β-Lactamase-Producing Enterobacterales in Municipal Wastewater Collections, Switzerland, 2019-2023. Emerg Infect Dis 2025; 31:574-578. [PMID: 40023816 PMCID: PMC11878307 DOI: 10.3201/eid3103.240099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025] Open
Abstract
We quantified presumptive extended-spectrum β-lactamase-producing Escherichia coli and Klebsiella, Enterobacter, Serratia, and Citrobacter group colonies from wastewater in Basel, Switzerland, across 3 years to represent before, during, and after the COVID-19 pandemic. Wastewater surveillance might be a noninvasive, sensitive, rapid, and cost-effective instrument for early detection and monitoring local epidemiology.
Collapse
Affiliation(s)
| | | | - Ana B. García-Martín
- University Hospital Basel, Basel, Switzerland (L. Aguilar-Bultet, E. Gómez-Sanz, Ana B. García-Martín, S. Tschudin-Sutter); State Laboratory Basel-City, Basel (M. Alt Hug, R. Furger, L. Eichenberger, C. Bagutti)
| | - Monica Alt Hug
- University Hospital Basel, Basel, Switzerland (L. Aguilar-Bultet, E. Gómez-Sanz, Ana B. García-Martín, S. Tschudin-Sutter); State Laboratory Basel-City, Basel (M. Alt Hug, R. Furger, L. Eichenberger, C. Bagutti)
| | - Reto Furger
- University Hospital Basel, Basel, Switzerland (L. Aguilar-Bultet, E. Gómez-Sanz, Ana B. García-Martín, S. Tschudin-Sutter); State Laboratory Basel-City, Basel (M. Alt Hug, R. Furger, L. Eichenberger, C. Bagutti)
| | - Lucas Eichenberger
- University Hospital Basel, Basel, Switzerland (L. Aguilar-Bultet, E. Gómez-Sanz, Ana B. García-Martín, S. Tschudin-Sutter); State Laboratory Basel-City, Basel (M. Alt Hug, R. Furger, L. Eichenberger, C. Bagutti)
| | | | | |
Collapse
|
6
|
Akintoyese TO, Alao JO, Oladipo EK, Oyedemi OT, Oyawoye OM. Antimicrobial resistance and virulence in Klebsiella pneumoniae: a four-month study in Osogbo, Nigeria. ANTIMICROBIAL STEWARDSHIP & HEALTHCARE EPIDEMIOLOGY : ASHE 2025; 5:e64. [PMID: 40026760 PMCID: PMC11869054 DOI: 10.1017/ash.2025.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 03/05/2025]
Abstract
Objective Antimicrobial resistance (AMR) is a growing global health crisis, with Klebsiella pneumoniae being a key pathogen due to its multidrug resistance (MDR). This study aimed to investigate the resistance profiles, demographic correlations, and molecular characteristics of MDR K. pneumoniae at UNIOSUN Teaching Hospital, Osogbo, Nigeria. Methods From January to April 2022, 99 clinical isolates (K. pneumoniae) were collected from various specimen types (blood, sputum, urine, wound, stool, and oral cavity). Antibiotic susceptibility was assessed using the Kirby-Bauer disk diffusion method, and virulence genes were analysed using multiplex polymerase chain reaction. Results All isolates exhibited resistance to ceftriaxone, cefotaxime, and colistin, with high resistance observed for cefepime and carbapenems (meropenem, imipenem, and ertapenem). Molecular characterization revealed the presence of virulence genes K1, K2, and mrkD in 15 isolates, while other tested virulence genes ( fimH, ramA, traT, K3, and K5 ) were not detected. Significant associations were identified between resistance patterns and demographic factors, including age and sex, highlighting potential vulnerabilities in specific populations. Conclusions This study underscores the alarming prevalence of MDR K. pneumoniae and aligns with global trends of rising AMR. Addressing these challenges requires targeted antimicrobial stewardship programs, infection control measures, public education, and enhanced surveillance systems. Incorporating molecular resistance testing and novel therapeutic agents in future research is crucial to developing effective containment strategies and preserving antibiotic efficacy.
Collapse
Affiliation(s)
| | - Jude Oluwapelumi Alao
- School of Public Health and Interdisciplinary Studies, Auckland University of Technology, Auckland, New Zealand
| | | | | | | |
Collapse
|
7
|
Elashkar E, Alfaraj R, El-Borady OM, Amer MM, Algammal AM, El-Demerdash AS. Novel silver nanoparticle-based biomaterials for combating Klebsiella pneumoniae biofilms. Front Microbiol 2025; 15:1507274. [PMID: 39850135 PMCID: PMC11754292 DOI: 10.3389/fmicb.2024.1507274] [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: 10/08/2024] [Accepted: 12/09/2024] [Indexed: 01/25/2025] Open
Abstract
Background Klebsiella pneumoniae is a significant nosocomial pathogen that has developed resistance to multiple antibiotics, often forming biofilms that enhance its virulence. This study investigated the efficacy of a novel nanoformulation, AgNPs@chitosan-NaF, in combating K. pneumoniae biofilms. Methods Antimicrobial susceptibility testing was performed to assess the antibiotic resistance profile of K. pneumoniae isolates. The antibiofilm activity of AgNPs@chitosan-NaF was evaluated using crystal violet staining and scanning electron microscopy. The underlying mechanisms of action were investigated through gene expression analysis. Results The majority of K. pneumoniae isolates exhibited high levels of multidrug resistance. AgNPs@chitosan-NaF demonstrated superior biofilm inhibition compared to AgNPs@chitosan, significantly reducing biofilm biomass and disrupting biofilm architecture at MICs ranging from 0.125 to 1 μg/mL. Mechanistic studies revealed that the nanoformulation downregulated the expression of key biofilm-associated genes, including treC, fimA, mrkA, and ecpA. While AgNPs@chitosan-NaF exhibited a concentration-dependent cytotoxic effect on both normal and cancer cell lines, minimal cytotoxicity was observed at concentrations below 31.25 μg/mL. Conclusion This study highlights the synergistic effect of silver nanoparticles, chitosan, and sodium fluoride in combating K. pneumoniae biofilms. The nanoformulation, AgNPs@chitosan-NaF, emerges as a promising therapeutic strategy to address the challenge of multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Eslam Elashkar
- Department of Botany and Microbiology, Faculty of Science, Benha University, Benha, Egypt
| | - Rihaf Alfaraj
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ola M. El-Borady
- Institute of Nanoscience and Nanotechnology, Kafrelsheikh University, Kafr ElSheikh, Egypt
| | - Mahmoud M. Amer
- Department of Botany and Microbiology, Faculty of Science, Benha University, Benha, Egypt
| | - Abdelazeem M. Algammal
- Department of Bacteriology, Immunology, and Mycology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Azza S. El-Demerdash
- Laboratory of Biotechnology, Department of Microbiology, Agricultural Research Center, Animal Health Research Institute, Zagazig, Egypt
| |
Collapse
|
8
|
Zhang M, Zhan M, Song X. Echinacoside attenuates Klebsiella pneumoniae-induced pneumonia via inhibition of the TLR4/NF-κB signaling. APMIS 2025; 133:e13507. [PMID: 39757690 DOI: 10.1111/apm.13507] [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] [Accepted: 12/03/2024] [Indexed: 01/07/2025]
Abstract
The Gram-negative bacterium Klebsiella pneumoniae (K. pneumoniae) is one major causative agent of community- and hospital-acquired pneumonia. Echinacoside (ECH) is a phenylethanoid glycoside isolated from Cistanche deserticola that possesses anti-inflammatory activity. Our research aimed to confirm whether ECH alleviates K. pneumoniae-induced pneumonia and explore the underlying regulatory mechanisms. BEAS-2B cells and BALB/c mice were infected by K. pneumoniae to establish the cellular and animal models, respectively, followed by ECH treatment. Inflammatory cytokine levels were detected by RT-qPCR and ELISA. The lung wet/dry (W/D) weight ratio and the myeloperoxidase (MPO) activity in lung tissues were examined. The pulmonary histopathologic changes were observed through hematoxylin and eosin (H&E) staining. The levels of TLR4/NF-κB pathway-associated molecules were estimated through western blotting, immunohistochemical, and immunohistochemical staining. K. pneumoniae infection caused lung histopathologic damage, enhanced MPO activity, elevated lung W/D weight ratio, and upregulated inflammatory cytokine levels in mice and promoted inflammatory cytokine expression in BEAS-2B cells, which were reversed by ECH treatment. K. pneumoniae infection-induced upregulation in TLR4, phosphorylated (p)-p65, and p-IκBα levels, and downregulation in IκBα levels in BEAS-2B cells and pneumonia mice were overturned by ECH treatment. ECH ameliorates K. pneumoniae-induced pneumonia through suppressing the TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Mi Zhang
- Department of Respiratory, Yichang Central People's Hospital, Yichang, China
| | - Ming Zhan
- Department of Respiratory, Yichang Central People's Hospital, Yichang, China
| | - Xinyu Song
- Department of Respiratory, Yichang Central People's Hospital, Yichang, China
| |
Collapse
|
9
|
Aggarwal M, Patra A, Awasthi I, George A, Gagneja S, Gupta V, Capalash N, Sharma P. Drug repurposing against antibiotic resistant bacterial pathogens. Eur J Med Chem 2024; 279:116833. [PMID: 39243454 DOI: 10.1016/j.ejmech.2024.116833] [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: 05/06/2024] [Revised: 08/22/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
The growing prevalence of MDR and XDR bacterial pathogens is posing a critical threat to global health. Traditional antibiotic development paths have encountered significant challenges and are drying up thus necessitating innovative approaches. Drug repurposing, which involves identifying new therapeutic applications for existing drugs, offers a promising alternative to combat resistant pathogens. By leveraging pre-existing safety and efficacy data, drug repurposing accelerates the development of new antimicrobial therapy regimes. This review explores the potential of repurposing existing FDA approved drugs against the ESKAPE and other clinically relevant bacterial pathogens and delves into the identification of suitable drug candidates, their mechanisms of action, and the potential for combination therapies. It also describes clinical trials and patent protection of repurposed drugs, offering perspectives on this evolving realm of therapeutic interventions against drug resistance.
Collapse
Affiliation(s)
- Manya Aggarwal
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Anushree Patra
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Ishita Awasthi
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Annu George
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Simran Gagneja
- Departmen of Microbiology, Panjab University, Chandigarh, India
| | - Varsha Gupta
- Department of Microbiology, Government Multi-speciality hospital, Sector 16, Chandigarh, India
| | - Neena Capalash
- Department of Biotechnology, Panjab University, Chandigarh, India
| | - Prince Sharma
- Departmen of Microbiology, Panjab University, Chandigarh, India.
| |
Collapse
|
10
|
Braun HG, Perera SR, Tremblay YD, Thomassin JL. Antimicrobial resistance in Klebsiella pneumoniae: an overview of common mechanisms and a current Canadian perspective. Can J Microbiol 2024; 70:507-528. [PMID: 39213659 DOI: 10.1139/cjm-2024-0032] [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] [Indexed: 09/04/2024]
Abstract
Klebsiella pneumoniae is a ubiquitous opportunistic pathogen of the family Enterobacteriaceae. K. pneumoniae is a member of the ESKAPEE pathogens (Enterococcus faecium, Staphylococcus aureus, K. pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter spp., and Escherichia coli), a group of bacteria that cause nosocomial infections and are able to resist killing by commonly relied upon antimicrobial agents. The acquisition of antimicrobial resistance (AMR) genes is increasing among community and clinical isolates of K. pneumoniae, making K. pneumoniae a rising threat to human health. In addition to the increase in AMR, K. pneumoniae is also thought to disseminate AMR genes to other bacterial species. In this review, the known mechanisms of K. pneumoniae AMR will be described and the current state of AMR K. pneumoniae within Canada will be discussed, including the impact of the coronavirus disease-2019 pandemic, current perspectives, and outlook for the future.
Collapse
Affiliation(s)
- Hannah G Braun
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Sumudu R Perera
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Yannick Dn Tremblay
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Jenny-Lee Thomassin
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
11
|
Huang J, Alhejaili AY, Alkherd UH, Milner M, Zhou G, Alzahrani D, Banzhaf M, Alzaidi AA, Rajeh AA, Al-Otaiby MA, Alabbad SS, Bukhari D, Aljurayan AN, Aljasham AT, Alzeyadi ZA, Alajel SM, Hong PY, Alghoribi M, Almutairi MM, Pain A, Salem WA, Moradigaravand D. The dissemination of multidrug-resistant and hypervirulent Klebsiella pneumoniae clones across the Kingdom of Saudi Arabia. Emerg Microbes Infect 2024; 13:2427793. [PMID: 39508718 PMCID: PMC11583321 DOI: 10.1080/22221751.2024.2427793] [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: 04/01/2024] [Revised: 10/14/2024] [Accepted: 11/06/2024] [Indexed: 11/15/2024]
Abstract
Klebsiella pneumoniae is a Gram-negative bacterium associated with a wide range of community- and hospital-acquired infections. The emergence of clonal hypervirulent strains resistant to last-resort antimicrobial agents has become a global concern. The Kingdom of Saudi Arabia (KSA), with its diverse population and high tourism traffic, serves as a platform where the spread of multidrug-resistant (MDR) strains are facilitated. However, the knowledge of epidemiology and population diversity of MDR K. pneumoniae in KSA is scarce. We conducted a comprehensive genomic survey on 352 MDR K. pneumoniae isolates systematically collected from bloodstream and urinary tract infections in 34 hospitals across 15 major cities in KSA during 2022 and 2023. Whole-genome sequencing on the isolates was performed, followed by genomic epidemiology and phylodynamic analysis. Our study revealed a dynamic population characterized by the rapid expansion of several dominant clones, including, ST2096, ST147, and ST231, which were estimated to have emerged within the past decade. These clones exhibited widespread dissemination across hospitals and were genetically linked to global strains, particularly from the Middle East and South Asia. All major clones harboured plasmid-borne ESBLs and carbapenemase genes, with plasmidome analysis identifying multiple IncH, IncA/C and IncL plasmids underlying the MDR-hypervirulent phenotype. These plasmids were shared between major clones and became acquired on the same time scales as the expansion of the dominant clones. Our results report ST2096 as an emerging MDR-hypervirulent clone, emphasizing the need for monitoring of the circulating clones and their plasmid content in the KSA and broader West Asia.
Collapse
Affiliation(s)
- Jiayi Huang
- Laboratory of Infectious Disease Epidemiology, KAUST Center of Excellence for Smart Health and Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Ahmed Yousef Alhejaili
- Ministry of Health, Riyadh, Kingdom of Saudi Arabia
- Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Mathew Milner
- Laboratory of Infectious Disease Epidemiology, KAUST Center of Excellence for Smart Health and Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Ge Zhou
- Laboratory of Infectious Disease Epidemiology, KAUST Center of Excellence for Smart Health and Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Deema Alzahrani
- Laboratory of Infectious Disease Epidemiology, KAUST Center of Excellence for Smart Health and Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| | - Manuel Banzhaf
- Newcastle University Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | | | | | | | | | - Doua Bukhari
- Ministry of Health, Riyadh, Kingdom of Saudi Arabia
| | | | - Alanoud T. Aljasham
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Zeyad A. Alzeyadi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqraa, Saudi Arabia
| | - Sulaiman M. Alajel
- Executive Department of Reference Laboratories, Research and Laboratories Sector, Saudi Food and Drug Authority, Riyadh, Saudi Arabia
| | - Pei-Ying Hong
- Environmental Science and Engineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | | | - Mashal M. Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Arnab Pain
- Pathogen Genomics Laboratory, KAUST Center of Excellence for Smart Health and Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Jeddah Makkah, Saudi Arabia
- International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | | | - Danesh Moradigaravand
- Laboratory of Infectious Disease Epidemiology, KAUST Center of Excellence for Smart Health and Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Kingdom of Saudi Arabia
| |
Collapse
|
12
|
Tsang KK, Lam MMC, Wick RR, Wyres KL, Bachman M, Baker S, Barry K, Brisse S, Campino S, Chiaverini A, Cirillo DM, Clark T, Corander J, Corbella M, Cornacchia A, Cuénod A, D'Alterio N, Di Marco F, Donado-Godoy P, Egli A, Farzana R, Feil EJ, Fostervold A, Gorrie CL, Hassan B, Hetland MAK, Hoa LNM, Hoi LT, Howden B, Ikhimiukor OO, Jenney AWJ, Kaspersen H, Khokhar F, Leangapichart T, Ligowska-Marzęta M, Löhr IH, Long SW, Mathers AJ, McArthur AG, Nagaraj G, Oaikhena AO, Okeke IN, Perdigão J, Parikh H, Pham MH, Pomilio F, Raffelsberger N, Rakotondrasoa A, Kumar KLR, Roberts LW, Rodrigues C, Samuelsen Ø, Sands K, Sassera D, Seth-Smith H, Shamanna V, Sherry NL, Sia S, Spadar A, Stoesser N, Sunde M, Sundsfjord A, Thach PN, Thomson NR, Thorpe HA, Torok ME, Trang VD, Trung NV, Vornhagen J, Walsh T, Warne B, Wilson H, Wright GD, Holt KE, KlebNET-GSP AMR Genotype-Phenotype Group. Diversity, functional classification and genotyping of SHV β-lactamases in Klebsiella pneumoniae. Microb Genom 2024; 10:001294. [PMID: 39432416 PMCID: PMC11493186 DOI: 10.1099/mgen.0.001294] [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: 04/08/2024] [Accepted: 08/23/2024] [Indexed: 10/23/2024] Open
Abstract
Interpreting the phenotypes of bla SHV alleles in Klebsiella pneumoniae genomes is complex. Whilst all strains are expected to carry a chromosomal copy conferring resistance to ampicillin, they may also carry mutations in chromosomal bla SHV alleles or additional plasmid-borne bla SHV alleles that have extended-spectrum β-lactamase (ESBL) activity and/or β-lactamase inhibitor (BLI) resistance activity. In addition, the role of individual mutations/a changes is not completely documented or understood. This has led to confusion in the literature and in antimicrobial resistance (AMR) gene databases [e.g. the National Center for Biotechnology Information (NCBI) Reference Gene Catalog and the β-lactamase database (BLDB)] over the specific functionality of individual sulfhydryl variable (SHV) protein variants. Therefore, the identification of ESBL-producing strains from K. pneumoniae genome data is complicated. Here, we reviewed the experimental evidence for the expansion of SHV enzyme function associated with specific aa substitutions. We then systematically assigned SHV alleles to functional classes (WT, ESBL and BLI resistant) based on the presence of these mutations. This resulted in the re-classification of 37 SHV alleles compared with the current assignments in the NCBI's Reference Gene Catalog and/or BLDB (21 to WT, 12 to ESBL and 4 to BLI resistant). Phylogenetic and comparative genomic analyses support that (i) SHV-1 (encoded by bla SHV-1) is the ancestral chromosomal variant, (ii) ESBL- and BLI-resistant variants have evolved multiple times through parallel substitution mutations, (iii) ESBL variants are mostly mobilized to plasmids and (iv) BLI-resistant variants mostly result from mutations in chromosomal bla SHV. We used matched genome-phenotype data from the KlebNET-GSP AMR Genotype-Phenotype Group to identify 3999 K. pneumoniae isolates carrying one or more bla SHV alleles but no other acquired β-lactamases to assess genotype-phenotype relationships for bla SHV. This collection includes human, animal and environmental isolates collected between 2001 and 2021 from 24 countries. Our analysis supports that mutations at Ambler sites 238 and 179 confer ESBL activity, whilst most omega-loop substitutions do not. Our data also provide support for the WT assignment of 67 protein variants, including 8 that were noted in public databases as ESBL. These eight variants were reclassified as WT because they lack ESBL-associated mutations, and our phenotype data support susceptibility to third-generation cephalosporins (SHV-27, SHV-38, SHV-40, SHV-41, SHV-42, SHV-65, SHV-164 and SHV-187). The approach and results outlined here have been implemented in Kleborate v2.4.1 (a software tool for genotyping K. pneumoniae), whereby known and novel bla SHV alleles are classified based on causative mutations. Kleborate v2.4.1 was updated to include ten novel protein variants from the KlebNET-GSP dataset and all alleles in public databases as of November 2023. This study demonstrates the power of sharing AMR phenotypes alongside genome data to improve the understanding of resistance mechanisms.
Collapse
Affiliation(s)
- Kara K. Tsang
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Margaret M. C. Lam
- Department of Infectious Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria 3004, Australia
| | - Ryan R. Wick
- Department of Infectious Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria 3004, Australia
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Kelly L. Wyres
- Department of Infectious Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria 3004, Australia
| | | | | | | | - Sylvain Brisse
- Institut Pasteur, Université Paris Cité, Biodiversity and Epidemiology of Bacterial Pathogens, Paris, France
| | - Susana Campino
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Alexandra Chiaverini
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| | | | - Taane Clark
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | | | - Marta Corbella
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alessandra Cornacchia
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| | - Aline Cuénod
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Nicola D'Alterio
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| | | | | | - Adrian Egli
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Refath Farzana
- Ineos-Oxford Institute for Antimicrobial Research, Department of Biology, University of Oxford, Oxford, UK
| | - Edward J. Feil
- The Milner Centre for Evolution, Department of Life Sciences, University of Bath, BA2 7AY, Bath, UK
| | - Aasmund Fostervold
- Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway
| | - Claire L. Gorrie
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | | - Le Thi Hoi
- Hanoi Medical University, Hanoi, Vietnam
| | - Benjamin Howden
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Odion O. Ikhimiukor
- Department of Pharmaceutical Microbiology, University of Ibadan, Ibadan, Nigeria
| | - Adam W. J. Jenney
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | | | | - Iren Høyland Löhr
- Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway
| | - Scott W. Long
- Houston Methodist, Weill Cornell Medical College, New York, USA
| | | | - Andrew G. McArthur
- Michael G. DeGroote Institute for Infectious Disease Research and Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Geetha Nagaraj
- Central Research Laboratory, Kempegowda Institute of Medical Sciences, Bengaluru, India
| | - Anderson O. Oaikhena
- Department of Pharmaceutical Microbiology, University of Ibadan, Ibadan, Nigeria
| | - Iruka N. Okeke
- Department of Pharmaceutical Microbiology, University of Ibadan, Ibadan, Nigeria
| | | | | | | | - Francesco Pomilio
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| | - Niclas Raffelsberger
- Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
| | | | - K. L. Ravi Kumar
- Central Research Laboratory, Kempegowda Institute of Medical Sciences, Bengaluru, India
| | | | - Carla Rodrigues
- Institut Pasteur, Université Paris Cité, Biodiversity and Epidemiology of Bacterial Pathogens, Paris, France
| | - Ørjan Samuelsen
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Kirsty Sands
- Ineos-Oxford Institute for Antimicrobial Research, Department of Biology, University of Oxford, Oxford, UK
| | - Davide Sassera
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Helena Seth-Smith
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Varun Shamanna
- Central Research Laboratory, Kempegowda Institute of Medical Sciences, Bengaluru, India
| | - Norelle L. Sherry
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Sonia Sia
- Research Institute for Tropical Medicine, Department of Health, Manila, Philippines
| | - Anton Spadar
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Nicole Stoesser
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Arnfinn Sundsfjord
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | | | | | | | | | | | | | - Jay Vornhagen
- Indiana University School of Medicine, Indianapolis, USA
| | - Timothy Walsh
- Ineos-Oxford Institute for Antimicrobial Research, Department of Biology, University of Oxford, Oxford, UK
| | - Ben Warne
- University of Cambridge, Cambridge, UK
| | - Hayley Wilson
- PHG Foundation, University of Cambridge, Cambridge, UK
| | - Gerard D. Wright
- Michael G. DeGroote Institute for Infectious Disease Research and Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Kathryn E. Holt
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
- Department of Infectious Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria 3004, Australia
| | - KlebNET-GSP AMR Genotype-Phenotype Group
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
- Department of Infectious Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria 3004, Australia
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- University of Michigan, Ann Arbor, USA
- University of Cambridge, Cambridge, UK
- University of Virginia, Charlottesville, USA
- Institut Pasteur, Université Paris Cité, Biodiversity and Epidemiology of Bacterial Pathogens, Paris, France
- Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
- Ospedale San Raffaele s.r.l. via olgettina, Milano, Italy
- University of Oslo, Oslo, Norway
- Microbiology and Virology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
- Centro de Investigación Tibaitatá de AGROSAVIA, Mosquera, Colombia
- Ineos-Oxford Institute for Antimicrobial Research, Department of Biology, University of Oxford, Oxford, UK
- The Milner Centre for Evolution, Department of Life Sciences, University of Bath, BA2 7AY, Bath, UK
- Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway
- Cardiff University, Cardiff, Wales, UK
- National Hospital for Tropical Diseases, Hanoi, Vietnam
- Hanoi Medical University, Hanoi, Vietnam
- Department of Pharmaceutical Microbiology, University of Ibadan, Ibadan, Nigeria
- Norwegian Veterinary Institute, Ås, Norway
- Statens Serum Institut, Copenhagen, Denmark
- Houston Methodist, Weill Cornell Medical College, New York, USA
- Michael G. DeGroote Institute for Infectious Disease Research and Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, Canada
- Central Research Laboratory, Kempegowda Institute of Medical Sciences, Bengaluru, India
- University of Lisbon, Lisbon, Portugal
- Wellcome Sanger Institute, Hinxton, UK
- Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
- Institut Pasteur de Bangui, Bangui, Central African Republic
- Queensland University of Technology, Brisbane, Australia
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
- Research Institute for Tropical Medicine, Department of Health, Manila, Philippines
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
- Indiana University School of Medicine, Indianapolis, USA
- PHG Foundation, University of Cambridge, Cambridge, UK
| |
Collapse
|
13
|
Zou P, Chen Z, Tu J, Chen X, Liu X. Comparison of Mutant Prevention Concentrations of Fluoroquinolones Against ESBL-Positive and ESBL-Negative Klebsiella pneumoniae Isolates from Orthopedic Patients. Microb Drug Resist 2024; 30:391-397. [PMID: 39019029 DOI: 10.1089/mdr.2024.0023] [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] [Indexed: 07/19/2024] Open
Abstract
The majority of Klebsiella pneumonia isolates possess the extended-spectrum beta-lactamase (ESBL) enzymes. Therefore, K. pneumoniae can easily develop drug resistance. How to effectively overcome the problem of drug resistance in K. pneumoniae is still a research hotspot. This study aimed to compare the mutant prevention concentration (MPC) of ESBL-positive and ESBL-negative K. pneumoniae isolated from orthopedic patients, which may provide a basis for the effective use of drugs to control the enrichment of resistance mutants of K. pneumoniae. The MPC90 values of 55 isolates of ESBL-positive K. pneumoniae against 4 fluoroquinolones were 32 µg/mL for levofloxacin and gatifloxacin, 16 µg/mL for ciprofloxacin, and 4 µg/mL for gemifloxacin. The selection index value was 8 for levofloxacin and ciprofloxacin and 2 for gemifloxacin and gatifloxacin, respectively. For ESBL-negative K. pneumoniae isolates, the MPC90 values were 16 µg/mL for levofloxacin and ciprofloxacin, 4 µg/mL for gemifloxacin, and 32 µg/mL for gatifloxacin. The selection index value was 8 for levofloxacin and ciprofloxacin, 2 for gemifloxacin, and 4 for gatifloxacin. For the ESBL-positive K. pneumoniae, the %T>MIC90 order was gemifloxacin > levofloxacin > ciprofloxacin > gatifloxacin. For the ESBL-negative K. pneumoniae, the %T>MIC90 order was levofloxacin > gemifloxacin > ciprofloxacin > gatifloxacin. The mutant-preventing ability of gatifloxacin and gemifloxacin was the strongest among the 4 fluoroquinolones. So gemifloxacin may be the first choice of drug to treat K. pneumoniae infection.
Collapse
Affiliation(s)
- Peng Zou
- The Department of Orthopedics, Zhengzhou Seventh People's Hospital, Zhengzhou, China
| | - Zhiquan Chen
- The Department of Orthopedics, Zhengzhou Seventh People's Hospital, Zhengzhou, China
| | - Jijun Tu
- The Department of Orthopedics, Zhengzhou Seventh People's Hospital, Zhengzhou, China
| | - Xinfeng Chen
- The Department of Orthopedics, Zhengzhou Seventh People's Hospital, Zhengzhou, China
| | - Xuejian Liu
- The Department of Orthopedics, Zhengzhou Seventh People's Hospital, Zhengzhou, China
| |
Collapse
|
14
|
Cardoso Almeida AP, de Moraes MA, da Silva AKF, Oliveira-Silva M, Nakamura-Silva R, de Almeida FM, Pappas Junior GJ, Pitondo-Silva A, de Campos TA. Long-term occurrence of multiple antimicrobial drug resistant Klebsiella pneumoniae isolates harboring virulent potential in a tertiary hospital from Brazil. Braz J Microbiol 2024; 55:2313-2320. [PMID: 38743244 PMCID: PMC11405615 DOI: 10.1007/s42770-024-01358-2] [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: 06/26/2023] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Klebsiella pneumoniae strains are globally associated with a plethora of opportunistic and severe human infections and are known to spread genes conferring antimicrobial resistance. Some strains harbor virulence determinants that enable them to cause serious disease in any patient, both in the hospital and in the community. The aim of this study was to determine the frequency of antimicrobial resistance and virulence traits (by gene detection and string test) among 83 K. pneumoniae isolates obtained from patient cultures of a scholar tertiary hospital in the Midwestern Brazil (Brasília, DF). Antimicrobial susceptibility analysis showed that 94% (78/83) of the isolates presented one of the following resistance profiles: resistant (R, 39), multidrug-resistant (MDR, 29), or extensively drug-resistant (XDR, 10). Several MDR and XDR strains harbored multiple virulence genes and displayed hypermucoviscous phenotype. These characteristics were observed among isolates obtained throughout all the sample collection period (2013 - 2017). The K2 serotype gene, a molecular marker of hypervirulence, was detected in three isolates, one of which classified as XDR. Sequence typing revealed the occurrence of isolates belonged to high-risk (ST13) and multiple resistance-spreading clones (ST105). Thus, our findings showed the occurrence of virulent potential isolates that also presented MDR/XDR phenotypes from 2013 to 2015. This study also indicates the probable convergence of virulence and resistance since at least 2013 in Brazil.
Collapse
Affiliation(s)
- Ana Paula Cardoso Almeida
- Programa de Pós-Graduação Em Biologia Microbiana, Universidade de Brasília, Brasília, Distrito Federal, Brasil
- Departamento de Biologia Celular, Instituto de Ciências Biológicas, Universidade de Brasília, Campus Universitário Darcy Ribeiro, Asa Norte, Brasilia, DF, CEP70910-900, Brazil
| | - Miguel Augusto de Moraes
- Faculdade de Ciências Farmacêuticas, Universidade de Ribeirão Preto, Ribeirão Preto, São Paulo, Brasil
- Programa de Pós-Graduação Em Tecnologia Ambiental, Universidade de Ribeirão Preto, Ribeirão Preto, São Paulo, Brasil
| | - Amanda Kamyla Ferreira da Silva
- Faculdade de Ciências Farmacêuticas, Universidade de Ribeirão Preto, Ribeirão Preto, São Paulo, Brasil
- Programa de Pós-Graduação Em Tecnologia Ambiental, Universidade de Ribeirão Preto, Ribeirão Preto, São Paulo, Brasil
| | - Mariana Oliveira-Silva
- Programa de Pós-Graduação Em Tecnologia Ambiental, Universidade de Ribeirão Preto, Ribeirão Preto, São Paulo, Brasil
| | - Rafael Nakamura-Silva
- Programa de Pós-Graduação Em Tecnologia Ambiental, Universidade de Ribeirão Preto, Ribeirão Preto, São Paulo, Brasil
| | | | - Georgios Joannis Pappas Junior
- Departamento de Biologia Celular, Instituto de Ciências Biológicas, Universidade de Brasília, Campus Universitário Darcy Ribeiro, Asa Norte, Brasilia, DF, CEP70910-900, Brazil
- Programa de Pós-Graduação Em Biologia Molecular, Universidade de Brasília, Brasília, DF, Brasil
| | - André Pitondo-Silva
- Faculdade de Ciências Farmacêuticas, Universidade de Ribeirão Preto, Ribeirão Preto, São Paulo, Brasil
- Programa de Pós-Graduação Em Tecnologia Ambiental, Universidade de Ribeirão Preto, Ribeirão Preto, São Paulo, Brasil
- Programa de Pós-Graduação Em Odontologia, Universidade de Ribeirão Preto, Ribeirão Preto, São Paulo, Brasil
| | - Tatiana Amabile de Campos
- Programa de Pós-Graduação Em Biologia Microbiana, Universidade de Brasília, Brasília, Distrito Federal, Brasil.
- Departamento de Biologia Celular, Instituto de Ciências Biológicas, Universidade de Brasília, Campus Universitário Darcy Ribeiro, Asa Norte, Brasilia, DF, CEP70910-900, Brazil.
| |
Collapse
|
15
|
Alshehri SM, Abdullah NS, Algarni A, AlZomia AS, Assiry MM. Resistance Pattern of Klebsiella pneumoniae in Aseer Region, Saudi Arabia: A Ten-Year Hospital-Based Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1344. [PMID: 39202625 PMCID: PMC11356269 DOI: 10.3390/medicina60081344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/11/2024] [Accepted: 08/18/2024] [Indexed: 09/03/2024]
Abstract
Background and Objectives: The frequency of multidrug-resistant Klebsiella pneumoniae (MDRKP) has dramatically increased worldwide in recent decades, posing an urgent threat to public health. The aim of this study was to assess the extent of K. pneumoniae in the Aseer region and explore the corresponding antimicrobial resistance profile over the last ten years. Materials and Methods: A record-based retrospective study was conducted in a tertiary hospital during the period of 2013 to 2022. The study targeted laboratory samples taken from patients admitted to the hospital and sent for K. pneumoniae culturing. We included only samples taken from the patient and confirmed by the lab. Data were extracted using a pre-structured data extraction sheet to avoid data-collection bias and confirm the inter-rater precision. Statistical Package for Social Sciences (SPSS) version 26 was employed for statistical analysis. All relationships were tested using Pearson X2 test for categorical data or chi-square for linear trend for resistance rate over years. Results: We obtained 3921 samples of isolated K. pneumoniae out of 28,420 bacterial samples. The isolation rate began at 11.3% in 2013, decreased to 6.1% in 2016, and then increased to a peak of 16.3% in 2021, before slightly decreasing to 12.8% in 2022. In total, 23.7% of K. pneumoniae samples were identified in urine samples, 19% in sputum samples, 14% in wound samples, and 11.7% in blood samples. The overall antibiotic resistance rate of K. pneumoniae from 2013 to 2022 showed a significant increase, particularly during 2020 and 2021, before decreasing again in 2022. The resistance rate decreased from 22.2% in 2013 to 18.6% in 2016 and increased to 54.6% and 56.4% during 2020 and 2021, respectively (p = 0.039). Conclusions: We observed a significant shift in K. pneumoniae resistance for some antibiotics during the study period, highlighting the urgent need for enhanced antimicrobial stewardship and infection-control measures.
Collapse
Affiliation(s)
- Saad Mohammed Alshehri
- Preventive Medicine and Public Health Physician, Ministry of Health, Abha 62515, Saudi Arabia
| | - Naif Saud Abdullah
- Community Medicine Consultant, Ministry of Health, Abha 62515, Saudi Arabia
| | - Abdullah Algarni
- Family Medicine Consultant, Aseer Central Hospital, Abha 62515, Saudi Arabia
| | | | - Mohammed Mushabub Assiry
- Senior Technician in the Department of Microbiology, Aseer Central Hospital, Abha 62515, Saudi Arabia
| |
Collapse
|
16
|
Fostervold A, Raffelsberger N, Hetland MAK, Bakksjø R, Bernhoff E, Samuelsen Ø, Sundsfjord A, Afset JE, Berntsen CF, Bævre-Jensen R, Ebbesen MH, Gammelsrud KW, Guleng AD, Handal N, Jakovljev A, Johal SK, Marvik Å, Natvik A, Sandnes RA, Tofteland S, Bjørnholt JV, Löhr IH. Risk of death in Klebsiella pneumoniae bloodstream infections is associated with specific phylogenetic lineages. J Infect 2024; 88:106155. [PMID: 38574775 DOI: 10.1016/j.jinf.2024.106155] [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: 01/31/2024] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Klebsiella pneumoniae species complex (KpSC) bloodstream infections (BSIs) are associated with considerable morbidity and mortality, particularly in elderly and multimorbid patients. Multidrug-resistant (MDR) strains have been associated with poorer outcome. However, the clinical impact of KpSC phylogenetic lineages on BSI outcome is unclear. METHODS In an 18-month nationwide Norwegian prospective study of KpSC BSI episodes in adults, we used whole-genome sequencing to describe the molecular epidemiology of KpSC, and multivariable Cox regression analysis including clinical data to determine adjusted hazard ratios (aHR) for death associated with specific genomic lineages. FINDINGS We included 1078 BSI episodes and 1082 bacterial isolates from 1055 patients. The overall 30-day case-fatality rate (CFR) was 12.5%. Median patient age was 73.4, 61.7% of patients were male. Median Charlson comorbidity score was 3. Klebsiella pneumoniae sensu stricto (Kp) (79.3%, n = 858/1082) and K. variicola (15.7%, n = 170/1082) were the dominating phylogroups. Global MDR-associated Kp clonal groups (CGs) were prevalent (25.0%, n = 270/1082) but 78.9% (n = 213/270) were not MDR, and 53.7% (n = 145/270) were community acquired. The major findings were increased risk for death within 30 days in monomicrobial BSIs caused by K. variicola (CFR 16.9%, n = 21; aHR 1.86, CI 1.10-3.17, p = 0.02), and global MDR-associated Kp CGs (CFR 17.0%, n = 36; aHR 1.52, CI 0.98-2.38, p = 0.06) compared to Kp CGs not associated with MDR (CFR 10.1%, n = 46). CONCLUSION Bacterial traits, beyond antimicrobial resistance, have a major impact on the clinical outcome of KpSC BSIs. The global spread of MDR-associated Kp CGs is driven by other mechanisms than antibiotic selection alone. Further insights into virulence determinants, and their association with phylogenetic lineages are needed to better understand the epidemiology of KpSC infection and clinical outcome.
Collapse
Affiliation(s)
- Aasmund Fostervold
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway; Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway.
| | - Niclas Raffelsberger
- Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway; Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Marit A K Hetland
- Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway; Department of Biological Sciences, Faculty of Mathematics and Natural Sciences, University of Bergen, Bergen, Norway
| | - Ragna Bakksjø
- Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway
| | - Eva Bernhoff
- Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway
| | - Ørjan Samuelsen
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway; Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Arnfinn Sundsfjord
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway; Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
| | - Jan E Afset
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Medical Microbiology, St. Olav's Hospital, Trondheim University hospital, Trondheim, Norway
| | - Christopher F Berntsen
- Department of Internal Medicine, Lovisenberg Diaconal Hospital, Oslo, Norway; Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Roar Bævre-Jensen
- Department of Medical Microbiology, Vestre Viken Hospital Trust, Drammen, Norway
| | - Marit H Ebbesen
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Karianne W Gammelsrud
- Department of Microbiology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anja D Guleng
- Centre for Laboratory Medicine, Østfold Hospital Trust, Kalnes, Norway
| | - Nina Handal
- Department of Microbiology and Infection control, Akershus University Hospital, Lørenskog, Norway
| | - Aleksandra Jakovljev
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Medical Microbiology, St. Olav's Hospital, Trondheim University hospital, Trondheim, Norway
| | - Simreen K Johal
- Department of Medical Microbiology, Nordland Hospital Trust, Bodø, Norway
| | - Åshild Marvik
- Department of Microbiology, Vestfold Hospital Trust, Tønsberg, Norway
| | - Ane Natvik
- Department of Medical Microbiology, Vestre Viken Hospital Trust, Bærum, Norway
| | - Rolf-Arne Sandnes
- Department of Medical Microbiology, Innlandet Hospital Trust, Lillehammer, Norway
| | - Ståle Tofteland
- Department of Microbiology, Hospital of Southern Norway Trust, Kristiansand, Norway
| | - Jørgen V Bjørnholt
- Department of Microbiology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Iren H Löhr
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway; Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
17
|
Fordham SME, Drobniewski F, Barrow M, Hutchings M, Crowther K, Richards D, Bolton P, Mantzouratou A, Sheridan E. Genetic Analyses of Rare ESBL ST628 Klebsiella pneumoniae Detected during a Protracted Nosocomial Outbreak in the United Kingdom. Microorganisms 2024; 12:883. [PMID: 38792715 PMCID: PMC11124425 DOI: 10.3390/microorganisms12050883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Klebsiella pneumoniae (K. pneumoniae) cultures from a hospital-wide outbreak in the UK, which lasted for over 12 months, were sequenced. We sought to sequence and genetically characterise the outbreak strain. Antibiotic Susceptibility Testing (AST) was performed on 65 K. pneumoniae isolates saved from the outbreak. All isolates were sequenced using the Oxford Nanopore Technologies (ONT) MinION flowcell: 10 isolates, including the isolate with the earliest collection date in 2017, were additionally sequenced on the NovaSeq 6000 platform to build high-accuracy nanopore-illumina assemblies. Among the sequenced strains, 60 were typed as ST628. 96.6% (n = 58/60) ST628 strains harboured a large ~247-kb FIB(K) plasmid carrying up to 11 antimicrobial resistance genes, including the extended-spectrum beta-lactamase (ESBL) gene, blaCTX-M-15. Clonality between the outbreak isolates was confirmed using single nucleotide polymorphism (SNP) typing. The outbreak strains were phylogenetically related to clinical ST628 strains identified in 2012, 6 years prior to the outbreak. A rare ESBL K. pneumoniae K2 ST628 strain harbouring a multi-drug resistant (MDR) plasmid encoding the ESBL gene blaCTX-M-15 was detected across multiple independent wards during the protracted nosocomial outbreak. Surveillance of this strain is recommended to prevent future nosocomial outbreaks.
Collapse
Affiliation(s)
- Stephen Mark Edward Fordham
- Department of Life & Environmental Sciences, Talbot Campus Fern Barrow, Bournemouth University, Poole BH12 5BB, UK; (S.M.E.F.); (M.B.); (A.M.)
| | - Francis Drobniewski
- Department of Infectious Diseases, Hammersmith Campus, Imperial College London, 8th Floor, Office 8.N10, DuCane Road, London W12 ONN, UK
- Department of Medical Microbiology, Poole Hospital, University Hospitals Dorset NHS Foundation Trust, Longfleet Road, Poole BH15 2JB, UK (P.B.)
| | - Magdalena Barrow
- Department of Life & Environmental Sciences, Talbot Campus Fern Barrow, Bournemouth University, Poole BH12 5BB, UK; (S.M.E.F.); (M.B.); (A.M.)
| | - Melissa Hutchings
- Department of Medical Microbiology, Poole Hospital, University Hospitals Dorset NHS Foundation Trust, Longfleet Road, Poole BH15 2JB, UK (P.B.)
| | - Kate Crowther
- Department of Medical Microbiology, Poole Hospital, University Hospitals Dorset NHS Foundation Trust, Longfleet Road, Poole BH15 2JB, UK (P.B.)
| | - Denise Richards
- Department of Medical Microbiology, Poole Hospital, University Hospitals Dorset NHS Foundation Trust, Longfleet Road, Poole BH15 2JB, UK (P.B.)
| | - Paul Bolton
- Department of Medical Microbiology, Poole Hospital, University Hospitals Dorset NHS Foundation Trust, Longfleet Road, Poole BH15 2JB, UK (P.B.)
| | - Anna Mantzouratou
- Department of Life & Environmental Sciences, Talbot Campus Fern Barrow, Bournemouth University, Poole BH12 5BB, UK; (S.M.E.F.); (M.B.); (A.M.)
| | - Elizabeth Sheridan
- Department of Medical Microbiology, Poole Hospital, University Hospitals Dorset NHS Foundation Trust, Longfleet Road, Poole BH15 2JB, UK (P.B.)
| |
Collapse
|
18
|
Hala S, Malaikah M, Huang J, Bahitham W, Fallatah O, Zakri S, Antony CP, Alshehri M, Ghazzali RN, Ben-Rached F, Alsahafi A, Alsaedi A, AlAhmadi G, Kaaki M, Alazmi M, AlhajHussein B, Yaseen M, Zowawi HM, Alghoribi MF, Althaqafi AO, Al-Amri A, Moradigaravand D, Pain A. The emergence of highly resistant and hypervirulent Klebsiella pneumoniae CC14 clone in a tertiary hospital over 8 years. Genome Med 2024; 16:58. [PMID: 38637822 PMCID: PMC11025284 DOI: 10.1186/s13073-024-01332-5] [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: 10/03/2023] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Klebsiella pneumoniae is a major bacterial and opportunistic human pathogen, increasingly recognized as a healthcare burden globally. The convergence of resistance and virulence in K. pneumoniae strains has led to the formation of hypervirulent and multidrug-resistant strains with dual risk, limiting treatment options. K. pneumoniae clones are known to emerge locally and spread globally. Therefore, an understanding of the dynamics and evolution of the emerging strains in hospitals is warranted to prevent future outbreaks. METHODS In this study, we conducted an in-depth genomic analysis on a large-scale collection of 328 multidrug-resistant (MDR) K. pneumoniae strains recovered from 239 patients from a single major hospital in the western coastal city of Jeddah in Saudi Arabia from 2014 through 2022. We employed a broad range of phylogenetic and phylodynamic methods to understand the evolution of the predominant clones on epidemiological time scales, virulence and resistance determinants, and their dynamics. We also integrated the genomic data with detailed electronic health record (EHR) data for the patients to understand the clinical implications of the resistance and virulence of different strains. RESULTS We discovered a diverse population underlying the infections, with most strains belonging to Clonal Complex 14 (CC14) exhibiting dominance. Specifically, we observed the emergence and continuous expansion of strains belonging to the dominant ST2096 in the CC14 clade across hospital wards in recent years. These strains acquired resistance mutations against colistin and extended spectrum β-lactamase (ESBL) and carbapenemase genes, namely blaOXA-48 and blaOXA-232, located on three distinct plasmids, on epidemiological time scales. Strains of ST2096 exhibited a high virulence level with the presence of the siderophore aerobactin (iuc) locus situated on the same mosaic plasmid as the ESBL gene. Integration of ST2096 with EHR data confirmed the significant link between colonization by ST2096 and the diagnosis of sepsis and elevated in-hospital mortality (p-value < 0.05). CONCLUSIONS Overall, these results demonstrate the clinical significance of ST2096 clones and illustrate the rapid evolution of an emerging hypervirulent and MDR K. pneumoniae in a clinical setting.
Collapse
Affiliation(s)
- Sharif Hala
- Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, 23955-6900, Jeddah, Makkah, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Ministry of National Guard Health Affairs, Riyadh, Western Region, Saudi Arabia
| | - Mohammed Malaikah
- Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, 23955-6900, Jeddah, Makkah, Saudi Arabia
- Laboratory of Infectious Disease Epidemiology, Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Jiayi Huang
- Laboratory of Infectious Disease Epidemiology, Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- KAUST Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Wesam Bahitham
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Ministry of National Guard Health Affairs, Riyadh, Western Region, Saudi Arabia
| | - Omniya Fallatah
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Ministry of National Guard Health Affairs, Riyadh, Western Region, Saudi Arabia
| | - Samer Zakri
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Ministry of National Guard Health Affairs, Riyadh, Western Region, Saudi Arabia
| | - Chakkiath Paul Antony
- Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, 23955-6900, Jeddah, Makkah, Saudi Arabia
- International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
| | - Mohammed Alshehri
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Ministry of National Guard Health Affairs, Riyadh, Western Region, Saudi Arabia
| | - Raeece Naeem Ghazzali
- Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, 23955-6900, Jeddah, Makkah, Saudi Arabia
| | - Fathia Ben-Rached
- Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, 23955-6900, Jeddah, Makkah, Saudi Arabia
| | - Abdullah Alsahafi
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Ministry of National Guard Health Affairs, Riyadh, Western Region, Saudi Arabia
| | - Asim Alsaedi
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Ministry of National Guard Health Affairs, Riyadh, Western Region, Saudi Arabia
| | - Ghadeer AlAhmadi
- King Faisal Specialist Hospital and Research Centre, Jeddah, Saudi Arabia
| | - Mai Kaaki
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Ministry of National Guard Health Affairs, Riyadh, Western Region, Saudi Arabia
| | - Meshari Alazmi
- KAUST Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- College of Computer Science and Engineering, University of Hail, Hail, Saudi Arabia
| | - Baraa AlhajHussein
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Ministry of National Guard Health Affairs, Riyadh, Western Region, Saudi Arabia
| | - Muhammad Yaseen
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Ministry of National Guard Health Affairs, Riyadh, Western Region, Saudi Arabia
| | - Hosam M Zowawi
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Ministry of National Guard Health Affairs, Riyadh, Western Region, Saudi Arabia
- The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, Australia
| | - Majed F Alghoribi
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Ministry of National Guard Health Affairs, Riyadh, Western Region, Saudi Arabia
| | - Abdulhakeem O Althaqafi
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Ministry of National Guard Health Affairs, Riyadh, Western Region, Saudi Arabia
| | - Abdulfattah Al-Amri
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Infectious Disease Research Department, King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Ministry of National Guard Health Affairs, Riyadh, Western Region, Saudi Arabia
| | - Danesh Moradigaravand
- Laboratory of Infectious Disease Epidemiology, Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia.
- KAUST Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia.
| | - Arnab Pain
- Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, 23955-6900, Jeddah, Makkah, Saudi Arabia.
- The University of Queensland, UQ Centre for Clinical Research, Herston, QLD, Australia.
| |
Collapse
|
19
|
Chirabhundhu N, Luk-In S, Phuadraksa T, Wichit S, Chatsuwan T, Wannigama DL, Yainoy S. Occurrence and mechanisms of tigecycline resistance in carbapenem- and colistin-resistant Klebsiella pneumoniae in Thailand. Sci Rep 2024; 14:5215. [PMID: 38433246 PMCID: PMC10909888 DOI: 10.1038/s41598-024-55705-2] [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: 01/10/2024] [Accepted: 02/27/2024] [Indexed: 03/05/2024] Open
Abstract
Tigecycline has been regarded as one of the most important last-resort antibiotics for the treatment of infections caused by extensively drug-resistant (XDR) bacteria, particularly carbapenem- and colistin-resistant Klebsiella pneumoniae (C-C-RKP). However, reports on tigecycline resistance have been growing. Overall, ~ 4000 K. pneumoniae clinical isolates were collected over a five-year period (2017-2021), in which 240 isolates of C-C-RKP were investigated. Most of these isolates (91.7%) were resistant to tigecycline. Notably, a high-risk clone of ST16 was predominantly identified, which was associated with the co-harboring of blaNDM-1 and blaOXA-232 genes. Their major mechanism of tigecycline resistance was the overexpression of efflux pump acrB gene and its regulator RamA, which was caused by mutations in RamR (M184V, Y59C, I141T, A28T, C99/C100 insertion), in RamR binding site (PI) of ramA gene (C139T), in MarR (S82G), and/or in AcrR (L154R, R13Q). Interestingly, four isolates of ST147 carried the mutated tet(A) efflux pump gene. To our knowledge, this is the first report on the prevalence and mechanisms of tigecycline resistance in C-C-RKP isolated from Thailand. The high incidence of tigecycline resistance observed among C-C-RKP in this study reflects an ongoing evolution of XDR bacteria against the last-resort antibiotics, which demands urgent action.
Collapse
Affiliation(s)
- Nachat Chirabhundhu
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Sirirat Luk-In
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Thanawat Phuadraksa
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Sineewanlaya Wichit
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Tanittha Chatsuwan
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in Antimicrobial Resistance and Stewardship, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Dhammika Leshan Wannigama
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in Antimicrobial Resistance and Stewardship, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
- School of Medicine, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, WA, Australia
- Biofilms and Antimicrobial Resistance Consortium of ODA Receiving Countries, The University of Sheffield, Sheffield, UK
- Pathogen Hunter's Research Collaborative Team, Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Sakda Yainoy
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand.
| |
Collapse
|
20
|
Verani JR, Blau DM, Gurley ES, Akelo V, Assefa N, Baillie V, Bassat Q, Berhane M, Bunn J, Cossa ACA, El Arifeen S, Gunturu R, Hale M, Igunza A, Keita AM, Kenneh S, Kotloff KL, Kowuor D, Mabunda R, Madewell ZJ, Madhi S, Madrid L, Mahtab S, Miguel J, Murila FV, Ogbuanu IU, Ojulong J, Onyango D, Oundo JO, Scott JAG, Sow S, Tapia M, Traore CB, Velaphi S, Whitney CG, Mandomando I, Breiman RF. Child deaths caused by Klebsiella pneumoniae in sub-Saharan Africa and south Asia: a secondary analysis of Child Health and Mortality Prevention Surveillance (CHAMPS) data. THE LANCET. MICROBE 2024; 5:e131-e141. [PMID: 38218193 PMCID: PMC10849973 DOI: 10.1016/s2666-5247(23)00290-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 07/25/2023] [Accepted: 08/30/2023] [Indexed: 01/15/2024]
Abstract
BACKGROUND Klebsiella pneumoniae is an important cause of nosocomial and community-acquired pneumonia and sepsis in children, and antibiotic-resistant K pneumoniae is a growing public health threat. We aimed to characterise child mortality associated with this pathogen in seven high-mortality settings. METHODS We analysed Child Health and Mortality Prevention Surveillance (CHAMPS) data on the causes of deaths in children younger than 5 years and stillbirths in sites located in seven countries across sub-Saharan Africa (Ethiopia, Kenya, Mali, Mozambique, Sierra Leone, and South Africa) and south Asia (Bangladesh) from Dec 9, 2016, to Dec 31, 2021. CHAMPS sites conduct active surveillance for deaths in catchment populations and following reporting of an eligible death or stillbirth seek consent for minimally invasive tissue sampling followed by extensive aetiological testing (microbiological, molecular, and pathological); cases are reviewed by expert panels to assign immediate, intermediate, and underlying causes of death. We reported on susceptibility to antibiotics for which at least 30 isolates had been tested, and excluded data on antibiotics for which susceptibility testing is not recommended for Klebsiella spp due to lack of clinical activity (eg, penicillin and ampicillin). FINDINGS Among 2352 child deaths with cause of death assigned, 497 (21%, 95% CI 20-23) had K pneumoniae in the causal chain of death; 100 (20%, 17-24) had K pneumoniae as the underlying cause. The frequency of K pneumoniae in the causal chain was highest in children aged 1-11 months (30%, 95% CI 26-34; 144 of 485 deaths) and 12-23 months (28%, 22-34; 63 of 225 deaths); frequency by site ranged from 6% (95% CI 3-11; 11 of 184 deaths) in Bangladesh to 52% (44-61; 71 of 136 deaths) in Ethiopia. K pneumoniae was in the causal chain for 450 (22%, 95% CI 20-24) of 2023 deaths that occurred in health facilities and 47 (14%, 11-19) of 329 deaths in the community. The most common clinical syndromes among deaths with K pneumoniae in the causal chain were sepsis (44%, 95% CI 40-49; 221 of 2352 deaths), sepsis in conjunction with pneumonia (19%, 16-23; 94 of 2352 deaths), and pneumonia (16%, 13-20; 80 of 2352 deaths). Among K pneumoniae isolates tested, 121 (84%) of 144 were resistant to ceftriaxone and 80 (75%) of 106 to gentamicin. INTERPRETATION K pneumoniae substantially contributed to deaths in the first 2 years of life across multiple high-mortality settings, and resistance to antibiotics used for sepsis treatment was common. Improved strategies are needed to rapidly identify and appropriately treat children who might be infected with this pathogen. These data suggest a potential impact of developing and using effective K pneumoniae vaccines in reducing neonatal, infant, and child deaths globally. FUNDING Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Jennifer R Verani
- Center for Global Health, US Centers for Disease Control and Prevention, Nairobi, Kenya.
| | - Dianna M Blau
- Center for Global Health, US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Emily S Gurley
- Maternal and Child Health Division, International Center for Diarrhoeal Diseases Research (icddr,b), Dhaka, Bangladesh; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Victor Akelo
- Center for Global Health, US Centers for Disease Control and Prevention Kenya, Kisumu, Kenya
| | - Nega Assefa
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Vicky Baillie
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Quique Bassat
- ISGlobal - Hospital Clínic, Universitat de Barcelona, Barcelona, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique; Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain; Hospital Sant Joan de Déu, Barcelona, Spain; Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Mussie Berhane
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - James Bunn
- World Health Organization, Sierra Leone, Freetown, Sierra Leone
| | - Anelsio C A Cossa
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Shams El Arifeen
- Maternal and Child Health Division, International Center for Diarrhoeal Diseases Research (icddr,b), Dhaka, Bangladesh
| | | | - Martin Hale
- National Health Laboratory Service, Department of Anatomical Pathology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Aggrey Igunza
- Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Adama M Keita
- Centre pour le Développement des Vaccins (CVD-Mali), Ministère de la Santé, Bamako, Mali
| | - Sartie Kenneh
- Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Karen L Kotloff
- Department of Pediatrics and Department of Medicine, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Rita Mabunda
- ISGlobal - Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Zachary J Madewell
- Center for Global Health, US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Shabir Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Lola Madrid
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia; Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Sana Mahtab
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Judice Miguel
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | | | | | | | | | - Joe O Oundo
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia; Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - J Anthony G Scott
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Samba Sow
- Centre pour le Développement des Vaccins (CVD-Mali), Ministère de la Santé, Bamako, Mali
| | - Milagritos Tapia
- Department of Pediatrics and Department of Medicine, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cheick B Traore
- Department of Pathological Anatomy and Cytology, University Hospital of Point G, Bamako, Mali
| | - Sithembiso Velaphi
- Department of Pediatrics, Chris Hani Baragwanath Academic Hospital, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Cynthia G Whitney
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Inacio Mandomando
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique; Instituto Nacional de Saúde (INS), Maputo, Mozambique
| | - Robert F Breiman
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA; Infectious Diseases and Oncology Research Institute, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
21
|
Rodríguez-Pallares S, Mateo-Vargas MA, Rodríguez-Iglesias MA, Galán-Sánchez F. Molecular characterization of consecutive isolates of OXA-48-producing Klebsiella pneumoniae: changes in the virulome using next-generation sequencing (NGS). Microbes Infect 2023; 25:105217. [PMID: 37716437 DOI: 10.1016/j.micinf.2023.105217] [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: 02/15/2023] [Revised: 09/09/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
Little is known about the clonality of consecutive OXA-48 producing-Klebsiella pneumoniae isolates from the same patient and the possibility of changes in their virulomes over time. We studied the molecular characteristics of twenty OXA-48-producing K. pneumoniae consecutive isolates from six patients using whole-genome sequencing. The genomes were screened for antimicrobial resistance and virulence factor genes and for replicon groups. MLST and SNPs analysis was performed. MLST analysis found 3 STs: ST11 (n = 13; 65.0%); ST4975 (n = 5, 25.0%); ST307 (n = 2; 10.0%). AcrAb efflux pump, siderophore enterobactin and rcsAB capsule synthesis regulator were detected in all sequenced isolates. The regulator of mucoid phenotype A (rmpA) and rmpA2 were not detected. Isolates also carried type 3 fimbriae (n = 19; 95.0%), yersiniabactin (n = 15; 75.0%) and type 1 fimbriae (7; 35.0%). Type 3 fimbriae and yersiniabactin were lost and recovered in consecutive isolates of two patients, probably acquired by horizontal gene transfer. Our findings reveal that recurrent infections are due to the same isolate, with an average of 2.69 SNPs per month, with different virulence profiles, and that the acquisition of virulence factor genes over time is possible.
Collapse
Affiliation(s)
- Salud Rodríguez-Pallares
- UGC Microbiología, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009, Cádiz, Spain.
| | | | - Manuel A Rodríguez-Iglesias
- UGC Microbiología, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009, Cádiz, Spain; Universidad de Cádiz, Cádiz, Spain; Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009, Cádiz, Spain
| | - Fátima Galán-Sánchez
- UGC Microbiología, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009, Cádiz, Spain; Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009, Cádiz, Spain
| |
Collapse
|
22
|
Oselusi SO, Sibuyi NRS, Meyer M, Madiehe AM. Ehretia Species Phytoconstituents as Potential Lead Compounds against Klebsiella pneumoniae Carbapenemase: A Computational Approach. BIOMED RESEARCH INTERNATIONAL 2023; 2023:8022356. [PMID: 37869630 PMCID: PMC10586912 DOI: 10.1155/2023/8022356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 09/05/2023] [Accepted: 09/26/2023] [Indexed: 10/24/2023]
Abstract
The evolution of antibiotic-resistant carbapenemase has negatively impacted the management of critical healthcare-associated infections. K. pneumoniae carbapenemase-2- (KPC-2-) expressing bacteria have developed resistance to conventional therapeutic options, including those used as a last resort for life-threatening diseases. In this study, Ehretia species phytoconstituents were screened for their potential to inhibit KPC-2 protein using in silico approaches. Molecular docking was used to identify strong KPC-2 protein binding phytoconstituents retrieved from the literature. The best-docked conformation of the ligands was selected based on their glide energy and binding interactions. To determine their binding free energies, these hit compounds were subjected to molecular mechanics with generalized born and surface area (MM-GBSA) in the PRIME module. Pharmacological assessments of the ligands were performed to evaluate their drug-likeness. Molecular dynamic (MD) simulations were used to analyze the conformational stability of the selected druglike compounds within the active site of the KPC-2 protein. Overall, a total of 69 phytoconstituents were compiled from the literature. Fourteen of these compounds exhibited a stronger binding affinity for the protein target than the reference drugs. Four of these top hit compounds, DB09, DB12, DB28, and DB66, revealed the highest efficacy in terms of drug-likeness properties. The MD simulation established that among the druglike compounds, DB66 attained stable conformations after 150 ns simulation in the active site of the protein. We concluded that DB66 from Ehretia species could play a significant role in therapeutic efforts against KPC-2-expressing bacteria.
Collapse
Affiliation(s)
- Samson O. Oselusi
- Nanobiotechnology Research Group, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa
- DSI/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa
| | - Nicole R. S. Sibuyi
- DSI/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa
| | - Mervin Meyer
- DSI/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa
| | - Abram M. Madiehe
- Nanobiotechnology Research Group, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa
- DSI/Mintek Nanotechnology Innovation Centre (NIC), Biolabels Research Node, Department of Biotechnology, University of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa
| |
Collapse
|
23
|
Centeleghe I, Norville P, Hughes L, Maillard JY. Klebsiella pneumoniae survives on surfaces as a dry biofilm. Am J Infect Control 2023; 51:1157-1162. [PMID: 36907360 DOI: 10.1016/j.ajic.2023.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 03/13/2023]
Abstract
BACKGROUND Dry surface biofilms (DSB) are widespread in healthcare settings presenting a challenge to cleaning and disinfection. Klebsiella pneumoniae has been a focus of attention due to antibiotic resistance and the emergence of hypervirulent strains. Few studies have demonstrated K pneumoniae survival on surfaces following desiccation. METHODS DSB were formed over 12 days. Bacterial culturability and transfer were investigated following DSB incubation up to 4 weeks. Bacterial viability in DSB was investigated with live/dead staining using flow cytometry. RESULTS K pneumoniae formed mature DSB. After 2 and 4 weeks of incubation, transfer from DSB was low (<55%) and reduced further (<21%) following wiping. Culturability at 2 and 4 weeks varied although viability remained high indicating viable but non culturable state (VBNC). DISCUSSION K pneumoniae was removed from surfaces by mechanical wiping as shown with DSB of other species. Although culturability was reduced over time, bacteria remained viable up to 4 weeks incubation, proving the need for robust cleaning regimens. CONCLUSIONS This is the first study confirming K pneumoniae survival on dry surfaces as a DSB. The presence of VBNC bacteria indicated that K pneumoniae can for extended periods, raising questions about its persistence on surfaces.
Collapse
Affiliation(s)
- Isabella Centeleghe
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, Cardiff, UK.
| | | | - Louise Hughes
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, Cardiff, UK
| | - Jean-Yves Maillard
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, Cardiff, UK
| |
Collapse
|
24
|
Qi Y, Xue JZ, Li SS, Elken EM, Haqmal MA, Li XS, Xu GY, Kong LC, Ma HX. Analysis of an IncR plasmid carried by carbapenem-resistant Klebsiella pneumoniae: A survey of swine Klebsiella pneumoniae in Jilin Province. J Glob Antimicrob Resist 2023; 34:83-90. [PMID: 37210003 DOI: 10.1016/j.jgar.2023.04.018] [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: 11/16/2022] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/22/2023] Open
Abstract
OBJECTIVES This study was conducted in Jilin Province to investigate the mechanism involved in the antibiotic resistance and pathogenicity of Klebsiella pneumoniae. METHODS Lung samples were collected from large-scale pig farms in Jilin Province. Antimicrobial susceptibility and mouse lethality assays were carried out. K. pneumoniae isolate JP20, with high virulence and antibiotic resistance, was chosen for whole-genome sequencing. The complete sequence of its genome was annotated, and the virulence and antibiotic resistance mechanism were analysed. RESULTS A total of 32 K. pneumoniae strains were isolated and tested for antibiotic resistance and pathogenicity. Among them, the JP20 strain showed high levels of resistance to all tested antimicrobial agents and strong pathogenicity in mice (lethal dose of 1.35 × 1011 CFU/mL). Sequencing of the multidrug-resistant and highly virulent K. pneumoniae JP20 strain revealed that the antibiotic resistance genes were mainly carried by an IncR plasmid. We speculate that extended-spectrum β-lactamases and loss of outer membrane porin OmpK36 play an important role in carbapenem antibiotic resistance. This plasmid contains a mosaic structure consisting of a large number of mobile elements. CONCLUSION Through genome-wide analysis, we found that an lncR plasmid carried by the JP20 strain may have evolved in pig farms, possibly leading to multidrug resistance in the JP20 strain. It is speculated that the antibiotic resistance of K. pneumoniae in pig farms is mainly mediated by mobile elements (insertion sequences, transposons, and plasmids). These data provide a basis for monitoring the antibiotic resistance of K. pneumoniae and lay a foundation for an improved understanding of the genomic characteristics and antibiotic resistance mechanism of K. pneumoniae.
Collapse
Affiliation(s)
- Yu Qi
- Department of Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Jun-Ze Xue
- Department of Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Shuang-Shuang Li
- Department of Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Emad Mohammed Elken
- Department of Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China; Department of Animal Production, Faculty of Agriculture, Al-Azhar University, Nasr City, Cairo, Egypt
| | - M Aman Haqmal
- Department of Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Xue-Song Li
- Department of Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Guan-Yi Xu
- Department of Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Ling-Cong Kong
- Department of Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China; The Key Laboratory of New Veterinary Drug Research and Development of Jilin Province, Jilin Agricultural University, Changchun, China.
| | - Hong-Xia Ma
- Department of Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China; The Key Laboratory of New Veterinary Drug Research and Development of Jilin Province, Jilin Agricultural University, Changchun, China; The Engineering Research Center of Bioreactor and Drug Development, Ministry of Education, Jilin Agricultural University, Changchun, China.
| |
Collapse
|
25
|
Raherison RE, Raharinavalona SA, Razanamparany T, Randrianotahiana TN, Randrianomanana TV, Andrianiaina MMA, Rakotomalala ADP, Andrianasolo RL. Urinary tract infection in diabetics hospitalized in Befelatanana Hospital, Antananarivo: Epidemiological, clinical, biological profiles and risk factors for multidrug-resistant bacterial infection. Clin Case Rep 2023; 11:e7867. [PMID: 37675415 PMCID: PMC10477472 DOI: 10.1002/ccr3.7867] [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: 06/12/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023] Open
Abstract
Key Clinical Message The main type of urinary tract infection in hospitalized diabetics in Antananarivo is acute pyelonephritis; Escherichia coli is the most isolated uropathogen; imipenem, amikacin, fosfomycin and ceftriaxone are the major antibiotics for which Escherichia coli retain good sensitivity; Type 2 diabetes is predictive factor for infection by multidrug resistant bacteria. Abstract This study aimed to describe the epidemiological-clinical profiles of diabetics hospitalized for bacterial urinary tract infections in the Endocrinology Department of Befelatanana Hospital, to identify the main bacteria responsible, their antibiotic sensitivity profile and the factors associated with multidrug-resistant bacterial infection. A cross-sectional study was conducted between March 2017 and March 2020 involving all diabetics hospitalized for documented community-acquired bacterial urinary tract infection during this period. The hospital prevalence of urinary tract infections was 4.64%. The mean age of the patients was 59.06 ± 14.26 years and the sex ratio was 0.15. The main sign was fever (55.76%). The main clinical form was uncomplicated acute pyelonephritis (38.46%). Fifty-seven bacterial uropathogens were isolated. The most frequent was Escherichia coli (77.19%). Escherichia coli was sensitive to ertapenem and nitrofurantoin in 100% of cases, to Amikacin in 97.5% of cases, to Fosfomycin in 94.4% of cases and to Ceftriaxone in 80.65% of cases. Thirteen patients were infected with multidrug-resistant bacteria, all of them are extended-spectrum beta-lactamase-producing Enterobacteriaceae. Only the type of diabetes was associated with multidrug-resistant bacteria infection. The epidemiological-clinical and biological characteristics of urinary tract infections in our diabetics are similar to those reported in the literature. Compliance with the rules of proper antibiotic use is imperative to limit the emergence and spread of multidrug-resistant bacteria.
Collapse
Affiliation(s)
- Rija Eric Raherison
- Endocrinology DepartmentJoseph Raseta Befelatanana University Hospital CenterAntananarivoMadagascar
| | | | - Thierry Razanamparany
- Endocrinology DepartmentJoseph Raseta Befelatanana University Hospital CenterAntananarivoMadagascar
| | | | | | | | | | | |
Collapse
|
26
|
M Shafik S, Abbas HA, Yousef N, Saleh MM. Crippling of Klebsiella pneumoniae virulence by metformin, N-acetylcysteine and secnidazole. BMC Microbiol 2023; 23:229. [PMID: 37608306 PMCID: PMC10464179 DOI: 10.1186/s12866-023-02969-9] [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: 06/08/2023] [Accepted: 08/04/2023] [Indexed: 08/24/2023] Open
Abstract
INTRODUCTION The emergence of multidrug-resistant Klebsiella pneumoniae in hospitals represents a serious threat to public health. Infections caused by Klebsiella pneumoniae are widespread in healthcare institutions, mainly pneumonia, bloodstream infections, and infections affecting neonates in intensive care units; so, it is necessary to combat this pathogen with new strategies. Targeting virulence factors necessary to induce host damage and disease is a new paradigm for antimicrobial therapy with several potential benefits that could lead to decreased resistance. BACKGROUND The influence of metformin, N-acetylcysteine, and secnidazole on Klebsiella pneumoniae virulence factors production was tested. The production of Klebsiella pneumoniae virulence factors such as biofilm formation, urease, proteases, hemolysins, and tolerance to oxidative stress was evaluated phenotypically using sub-inhibitory concentration (1/8 MIC) of metformin, N-acetylcysteine, and secnidazole. For more confirmation, qRT-PCR was used to assess the relative expression level of rmpA, wcaG, fimH-1, mrkD, ureA, and khe genes regulating virulence factors production. RESULTS Metformin, N-acetylcysteine, and secnidazole were all found to have a powerful inhibitory effect on the production of virulence factors phenotypically. Our results showed a significant reduction in the expression level of rmpA, wcaG, fimH-1, mrkD, ureA, and khe genes. Furthermore, the tested drugs were investigated in vivo to inform their ability to protect mice against Klebsiella pneumoniae pathogenesis. CONCLUSIONS Metformin, N-acetylcysteine, and secnidazole inhibited the virulence of Klebsiella pneumoniae. Besides combating resistant Klebsiella pneumoniae, the tested drugs could also serve as an adjuvant to traditional antibiotics.
Collapse
Affiliation(s)
- Shokri M Shafik
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig City, Egypt
| | - Hisham A Abbas
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig City, Egypt
| | - Nehal Yousef
- Microbiology and Immunology Department, Faculty of Pharmacy, Zagazig University, Zagazig City, Egypt
| | - Moustafa M Saleh
- Microbiology and Immunology Department, Faculty of Pharmacy, Port Said University, Port Said City, Egypt.
| |
Collapse
|
27
|
Dallman TJ, Neuert S, Fernandez Turienzo C, Berin M, Richardson E, Fuentes-Utrilla P, Loman N, Gharbia S, Jenkins C, Behrens RH, Godbole G, Brown M. Prevalence and Persistence of Antibiotic Resistance Determinants in the Gut of Travelers Returning to the United Kingdom is Associated with Colonization by Pathogenic Escherichia coli. Microbiol Spectr 2023; 11:e0518522. [PMID: 37255437 PMCID: PMC10433802 DOI: 10.1128/spectrum.05185-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 05/11/2023] [Indexed: 06/01/2023] Open
Abstract
The gut microbiota constitutes an ideal environment for the selection, exchange, and carriage of antibiotic resistance determinants (ARDs), and international travel has been identified as a risk factor for acquisition of resistant organisms. Here, we present a longitudinal metagenomic analysis of the gut resistome in travellers to "high-risk" countries (Gutback). Fifty volunteers, recruited at a travel clinic in London, United Kingdom, provided stool samples before (pre-travel), immediately after (post-travel), and 6 months after their return (follow-up) from a high-risk destination. Fecal DNA was extracted, metagenomic sequencing performed and the resistome profiled. An increase in abundance and diversity of resistome was observed after travel. Significant increases in abundance were seen in antimicrobial genes conferring resistance to macrolides, third-generation cephalosporins, aminoglycosides, and sulfonamides. There was a significant association with increased resistome abundance if the participant experienced diarrhea during travel or took antibiotics, but these two variables were co-correlated. The resistome abundance returned to pre-travel levels by the 6-month sample point but there was evidence of persistence of several ARDs. The post-travel samples had an increase in abundance Escherichia coli which was positively associated with many acquired resistant determinants. Virulence and phylogenetic profiling revealed pathogenic E. coli significantly contributed to this increase abundance. In summary, in this study, foreign travel remains a significant risk factor for acquisition of microbes conferring resistance to multiple classes of antibiotics, often associated with symptomatic exposure to diarrhoeagenic E. coli. IMPORTANCE A future where antimicrobial therapy is severely compromised by the increase in resistant organisms is of grave concern. Given the variability in prevalence and diversity of antimicrobial resistance determinants in different geographical settings, international travel is a known risk factor for acquisition of resistant organisms into the gut microbiota. In this study, we show the utility of metagenomic approaches to quantify the levels of acquisition and carriage of resistance determinants after travel to a "high-risk" setting. Significant modulation to the resistome was seen after travel that is largely resolved within 6 months, although evidence of persistence of several ARDs was observed. Risk factors for acquisition included experiencing a diarrheal episode and the use of antibiotics. Colonization by pathogenic Escherichia coli was correlated with an increase in acquisition of antimicrobial resistance determinants, and as such established public health guidance to travelers on food and water safety remain an important message to reduce the spread of antibiotic resistance.
Collapse
Affiliation(s)
- Timothy J. Dallman
- Gastrointestinal Bacteria Reference Unit, UK Health Security Agency London, United Kingdom
- Faculty of Veterinary Medicine, Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| | - Saskia Neuert
- Gastrointestinal Bacteria Reference Unit, UK Health Security Agency London, United Kingdom
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, United Kingdom
- National Institute for Health Research Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool, United Kingdom
| | - Cristina Fernandez Turienzo
- Hospital for Tropical Diseases, University College London Hospitals NHS Foundation Trust, London, United Kingdom
- Division of Infection and Immunity, University College London, London, United Kingdom
- Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Michelle Berin
- Hospital for Tropical Diseases, University College London Hospitals NHS Foundation Trust, London, United Kingdom
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Emily Richardson
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
- MicrobesNG, Birmingham, United Kingdom
| | - Pablo Fuentes-Utrilla
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
- MicrobesNG, Birmingham, United Kingdom
| | - Nicholas Loman
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Saheer Gharbia
- Gastrointestinal Bacteria Reference Unit, UK Health Security Agency London, United Kingdom
- National Institute for Health Research Health Protection Research Unit in Genomics and Enabling Data, Warwick University, United Kingdom
| | - Claire Jenkins
- Gastrointestinal Bacteria Reference Unit, UK Health Security Agency London, United Kingdom
- National Institute for Health Research Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool, United Kingdom
| | - Ron H. Behrens
- Hospital for Tropical Diseases, University College London Hospitals NHS Foundation Trust, London, United Kingdom
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Gauri Godbole
- Gastrointestinal Bacteria Reference Unit, UK Health Security Agency London, United Kingdom
- Hospital for Tropical Diseases, University College London Hospitals NHS Foundation Trust, London, United Kingdom
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Michael Brown
- Hospital for Tropical Diseases, University College London Hospitals NHS Foundation Trust, London, United Kingdom
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
28
|
He W, Wu C, Chen G, Zhang G, Zhao Z, Wen S, Zhou Y, Deng X, Feng Y, Zhong LL, Tian GB, Dai M. Comparative Genomic Analysis of Hypervirulence Carbapenem-Resistant Klebsiella pneumoniae from Inpatients with Infection and Gut Colonization, China. Infect Drug Resist 2023; 16:5251-5261. [PMID: 37601558 PMCID: PMC10437719 DOI: 10.2147/idr.s416770] [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: 05/25/2023] [Accepted: 07/12/2023] [Indexed: 08/22/2023] Open
Abstract
Background The emergence and spread of hypervirulent carbapenem-resistant Klebsiella pneumoniae (hv-CRKP) is a potential epidemiological threat that needs to be monitored. However, the transmission and pathogenic characteristics of hv-CRKP in China remain unclear. We investigated the epidemiological characteristics of gut colonized hv-CRKP in a hospital in Guangdong Province, China. Methods A total of 46 gut colonized hv-CRKP isolates were collected from Sun Yat-Sen Memorial Hospital (Guangzhou, China) from August 31st to December 31st, 2021. Minimum inhibitory concentrations (MICs) were obtained for 15 antibiotics for 46 hv-CRKP isolates. BALB/C mice infection model and mucoviscosity assay was used to evaluate the virulence of the isolates. The characteristics of genome, phylogenetic relationship and the structure of the plasmid of 46 gut colonized hv-CRKP isolates were compared with pathogenic isolates from GeneBank based on whole-genome data. Results The hv-CRKP isolation rate of all gut colonized carbapenem-resistant Klebsiella pneumoniae was 17% (46/270), and the intestinal colonization rate of hv-CRKP was irrelevant to the sex, age, department of hospitalization, and history of antibiotic use of the host. The gut colonized hv-CRKP showed pandrug resistance and hypervirulence. The gut colonized hv-CRKP and pathogenic hv-CRKP prevalent in China were mainly ST11 hv-CRKP and had two major epidemic clades. The similarities in genomic characteristics between gut colonized hv-CRKP and pathogenic hv-CRKP were consistent. The gut colonized hv-CRKP carried an incomplete structure pK2044 virulence plasmid from hypervirulent K. pneumoniae NTUH-K2044 by analyzing the virulence plasmid structure. Conclusion Our results suggest that the gut colonized ST11 hv-CRKP may serve as a reservoir for the clinical pathogenic ST11 HV-CRKP. It is necessary to further strengthen the monitoring of gut colonized hv-CRKP and research the potential mechanism of infection caused by gut colonized hv-CRKP.
Collapse
Affiliation(s)
- Wan He
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, 610500, People’s Republic of China
| | - Changbu Wu
- Department of Immunology and Microbiology, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
- Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, People’s Republic of China
| | - Guanping Chen
- Department of Immunology and Microbiology, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
- Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, People’s Republic of China
- Department of Immunology, School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, People’s Republic of China
| | - Guili Zhang
- Department of Immunology and Microbiology, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
- Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, People’s Republic of China
| | - Zihan Zhao
- Department of Immunology and Microbiology, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
- Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, People’s Republic of China
| | - Shu’an Wen
- Department of Immunology and Microbiology, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
- Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, People’s Republic of China
| | - Yuan Zhou
- Department of Immunology and Microbiology, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
- Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, People’s Republic of China
| | - Xue Deng
- Department of Immunology and Microbiology, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
- Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, People’s Republic of China
| | - Yu Feng
- Department of Immunology and Microbiology, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
- Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, People’s Republic of China
| | - Lan-Lan Zhong
- Department of Immunology and Microbiology, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
- Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, People’s Republic of China
| | - Guo-Bao Tian
- Department of Immunology and Microbiology, Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
- Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, People’s Republic of China
- Department of Immunology, School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, People’s Republic of China
| | - Min Dai
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, 610500, People’s Republic of China
| |
Collapse
|
29
|
Aruhomukama D, Nakabuye H. Investigating the evolution and predicting the future outlook of antimicrobial resistance in sub-saharan Africa using phenotypic data for Klebsiella pneumoniae: a 12-year analysis. BMC Microbiol 2023; 23:214. [PMID: 37553587 PMCID: PMC10408162 DOI: 10.1186/s12866-023-02966-y] [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: 05/25/2023] [Accepted: 08/01/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Antimicrobial resistance (AMR) is a major public health challenge, particularly in sub-Saharan Africa (SSA). This study aimed to investigate the evolution and predict the future outlook of AMR in SSA over a 12-year period. By analysing the trends and patterns of AMR, the study sought to enhance our understanding of this pressing issue in the region and provide valuable insights for effective interventions and control measures to mitigate the impact of AMR on public health in SSA. RESULTS The study found that general medicine patients had the highest proportion of samples with AMR. Different types of samples showed varying levels of AMR. Across the studied locations, the highest resistance was consistently observed against ceftaroline (ranging from 68 to 84%), while the lowest resistance was consistently observed against ceftazidime avibactam, imipenem, meropenem, and meropenem vaborbactam (ranging from 92 to 93%). Notably, the predictive analysis showed a significant increasing trend in resistance to amoxicillin-clavulanate, cefepime, ceftazidime, ceftaroline, imipenem, meropenem, piperacillin-tazobactam, and aztreonam over time. CONCLUSIONS These findings suggest the need for coordinated efforts and interventions to control and prevent the spread of AMR in SSA. Targeted surveillance based on local resistance patterns, sample types, and patient populations is crucial for effective monitoring and control of AMR. The study also highlights the urgent need for action, including judicious use of antibiotics and the development of alternative treatment options to combat the growing problem of AMR in SSA.
Collapse
Affiliation(s)
- Dickson Aruhomukama
- Department of Medical Microbiology, College of Health Sciences, Makerere University, Kampala, Uganda.
| | - Hellen Nakabuye
- Department of Medical Microbiology, College of Health Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
30
|
Pruss A, Kwiatkowski P, Sienkiewicz M, Masiuk H, Łapińska A, Kot B, Kilczewska Z, Giedrys-Kalemba S, Dołęgowska B. Similarity Analysis of Klebsiella pneumoniae Producing Carbapenemases Isolated from UTI and Other Infections. Antibiotics (Basel) 2023; 12:1224. [PMID: 37508320 PMCID: PMC10376303 DOI: 10.3390/antibiotics12071224] [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: 06/25/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Klebsiella pneumoniae is an important opportunistic pathogen responsible for severe infections, mainly urinary tract infections (UTIs) and pneumonia. Hospital epidemic infections caused by multiresistant strains of carbapenemase-producing K. pneumoniae are the most concerning. NDM-producing strains are resistant to a wide range of antibiotics and have become the most significant threat. Determining the natural reservoirs and routes of infections is essential to end hospital outbreaks. Understanding the relatedness of K. pneumoniae strains is essential to determine the range and nature of the infection. The study compared phylogenetic relatedness between multiresistant K. pneumoniae strains isolated from hospitalized patients. Susceptibility to drugs and mechanisms of resistance were confirmed using phenotypic methods. PFGE was used to analyze the relatedness between strains. We analyzed 69 K. pneumoniae strains from various healthcare units. The isolates were mainly identified from urine. Strains were resistant to β-lactam antibiotics with β-lactamase inhibitors, cephalosporins, and quinolones. Their susceptibility to aminoglycosides and carbapenem antibiotics was diverse. Most of the isolated strains produced New Delhi metallo-ß-lactamase (NDM). Although K. pneumoniae strains were classified into several genotype clusters, closely related isolates were confirmed in the same hospital's wards, and in two hospitals in the same province.
Collapse
Affiliation(s)
- Agata Pruss
- Department of Laboratory Medicine, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Paweł Kwiatkowski
- Department of Diagnostic Immunology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Monika Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostic, Medical University of Lodz, Muszynskiego St. 1, 90-151 Lodz, Poland
| | - Helena Masiuk
- Department of Medical Microbiology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Agnieszka Łapińska
- Department of Laboratory Medicine, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Barbara Kot
- Institute of Biological Sciences, Faculty of Exact and Natural Sciences, Siedlce University of Natural Sciences and Humanities, 14 Bolesława Prusa Str., 08-110 Siedlce, Poland
| | - Zuzanna Kilczewska
- Department of Laboratory Medicine, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Stefania Giedrys-Kalemba
- Department of Medical Microbiology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| | - Barbara Dołęgowska
- Department of Laboratory Medicine, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland
| |
Collapse
|
31
|
Yang M, Zhou X, Bao Y, Zhang Y, Liu B, Gan L, Tao W, Tuo J, Gong H. Comprehensive Genomic Analysis Reveals Extensive Diversity of Type I and Type IV Secretion Systems in Klebsiella pneumoniae. Curr Microbiol 2023; 80:270. [PMID: 37402963 DOI: 10.1007/s00284-023-03362-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 06/02/2023] [Indexed: 07/06/2023]
Abstract
The diversity and distribution of secretion systems in Klebsiella pneumoniae are unclear. In this study, the six common secretion systems (T1SS-T6SS) were comprehensively investigated in the genomes of 952 K. pneumoniae strains. T1SS, T2SS, type T subtype of T4SS, T5SS, and subtype T6SSi of T6SS were found. The findings indicated fewer types of secretion systems in K. pneumoniae than reported in Enterobacteriaceae, such as Escherichia coli. One conserved T2SS, one conserved T5SS, and two conserved T6SS were detected in more than 90% of the strains. In contrast, the strains displayed extensive diversity of T1SS and T4SS. Notably, T1SS and T4SS were enriched in the hypervirulent and classical multidrug resistance pathotypes of K. pneumoniae, respectively. The results expand the epidemiological knowledge of the virulence and transmissibility of pathogenic K. pneumoniae and contribute to identify the potential strains for safe applications.
Collapse
Affiliation(s)
- Menglei Yang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, People's Republic of China
| | - Xiaoqin Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, People's Republic of China
| | - Yangyang Bao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, People's Republic of China
| | - Yang Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, People's Republic of China
| | - Boya Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, People's Republic of China
| | - Luxi Gan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, People's Republic of China
| | - Weihua Tao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, People's Republic of China
| | - Jinyou Tuo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, People's Republic of China
| | - Heng Gong
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, People's Republic of China.
| |
Collapse
|
32
|
Mączyńska B, Frej-Mądrzak M, Sarowska J, Woronowicz K, Choroszy-Król I, Jama-Kmiecik A. Evolution of Antibiotic Resistance in Escherichia coli and Klebsiella pneumoniae Clinical Isolates in a Multi-Profile Hospital over 5 Years (2017-2021). J Clin Med 2023; 12:jcm12062414. [PMID: 36983414 PMCID: PMC10058544 DOI: 10.3390/jcm12062414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/10/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
In recent years, we have witnessed a growing drug resistance among bacteria, which is associated with the use and availability of an increasing number of broad-spectrum antimicrobial agents, as well as with their irrational and excessive use. The present study aims to analyze changes in the drug resistance of Gram-negative Enterobacterales: Escherichia coli and Klebsiella pneumoniae, isolated from infections in a multi-profile hospital over five years (from 2017 to 2021). Among the practical outcomes of the evaluation of these data will be the possibility of determining changes in susceptibility to the antibiotics used in the hospital. In turn, this will help propose new therapeutic options, especially for empirical therapy that is necessary in severe infections. The analysis of the use of individual groups of antibiotics allowed for identification of the causes of the increasing resistance of Gram-negative bacilli. The highest number of infections whose etiological agent was K. pneumoniae ESBL(+) and E. coli ESBL(+) was observed in 2018. In the analyzed five-year period, the number of multi-resistant (MDR) K. pneumoniae strains increased successively, which seems to be related to the growing use, especially in the pandemic period, of broad-spectrum antibiotics, mainly penicillins with inhibitors, third-generation cephalosporins, and carbapenems.
Collapse
Affiliation(s)
- Beata Mączyńska
- Department of Pharmaceutical Microbiology and Parasitology, Faculty of Pharmacy, Medical University, 50-367 Wroclaw, Poland
- Department of Hygiene and Epidemiology, Lower Silesian T. Marciniak Specialist Hospital-Center for Emergency Medicine, 54-049 Wrocław, Poland
| | - Magdalena Frej-Mądrzak
- Department of Basic Sciences, Faculty of Health Sciences, Medical University, 50-367 Wroclaw, Poland
| | - Jolanta Sarowska
- Department of Basic Sciences, Faculty of Health Sciences, Medical University, 50-367 Wroclaw, Poland
| | | | - Irena Choroszy-Król
- Department of Basic Sciences, Faculty of Health Sciences, Medical University, 50-367 Wroclaw, Poland
| | - Agnieszka Jama-Kmiecik
- Department of Basic Sciences, Faculty of Health Sciences, Medical University, 50-367 Wroclaw, Poland
| |
Collapse
|
33
|
Li S, Wu J, Ma N, Liu W, Shao M, Ying N, Zhu L. Prediction of genome-wide imipenem resistance features in Klebsiella pneumoniae using machine learning. J Med Microbiol 2023; 72. [PMID: 36753438 DOI: 10.1099/jmm.0.001657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Introduction. The resistance rate of Klebsiella pneumoniae (K. pneumoniae) to imipenem is increasing year by year, and the imipenem resistance mechanism of K. pneumoniae is complex. Therefore, it is urgent to develop new strategies to explore the resistance mechanism of imipenem for its effective and accurate use in clinical practice.Hypothesis/Gap sStatement. Machine learning could identify resistance features and biological process that influence microbial resistance from whole-genome sequencing (WGS) data.Aims. This work aimed to predict imipenem resistance genetic features in K. pneumoniae from whole-genome k-mer features, and analyse their function for understanding its resistance mechanism.Methods. This study analysed WGS data of K. pneumoniae combined with resistance phenotype for imipenem, and established K. pneumoniae to imipenem genotype-phenotype model to predict resistance features using chi-squared test and random forest. An external clinical dataset was used to verify prediction power of resistance features. The potential genes were identified through alignment the resistance features with the K. pneumoniae reference genome using blastn, the functions of potential genes were further analysed to explore its resistance-related signalling pathways with GO and KEGG analysis, the resistance sequence patterns were screened using streme software. Finally, the resistance features were combined and modelled through four machine-learning algorithms (logistic regression, SVM, GBDT and XGBoost) to evaluate their phenotype prediction ability.Results. A total of 16 670 imipenem resistance features were predicted from genotype-phenotype model. The 30 potential genes were identified by annotating the resistance features and corresponded to known antibiotic-related genes (mdtM, dedA, rne, etc.). GO and KEGG pathway analyses indicated the possible association of imipenem resistance with metabolism process and cell membrane. CRYCAGCDN and CGRDAAAN were found from the imipenem resistance features, which were widely presented in the reported β-lactam resistance genes (bla SHV, bla CTX-M, bla TEM, etc.), and YCYAGCMCAST with metabolic functions (organic substance metabolic process, nitrogen compound metabolic process and cellular metabolic process) was identified from the top 50 resistance features. The 25 resistance genes in the training dataset included 19 genes in the external dataset, which verified the accuracy of prediction. The area under curve values of logistics regression, SVM, GBDT and XGBoost were 0.965, 0.966, 0.969 and 0.969, respectively, indicating that the imipenem resistance features have a strong prediction power.Conclusion. Machine-learning methods could effectively predict the imipenem resistance feature in K. pneumoniae, and provide resistance sequence profiles for predicting resistance phenotype and exploring potential resistance mechanisms. It provides an important insight into the potential therapeutic strategies of K. pneumoniae resistance to imipenem, and speed up the application of machine learning in routine diagnosis.
Collapse
Affiliation(s)
- Shanshan Li
- College of Automation, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China
| | - Jun Wu
- Lin'an Center for Disease Control and Prevention, Lin'an, 311300, PR China
| | - Nan Ma
- College of Automation, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China
| | - Wenjia Liu
- College of Automation, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China.,College of Electronics and Information Engineering, Hangzhou Dianzi University, Hangzhou 310018, PR China
| | - Mengjie Shao
- College of Automation, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China
| | - Nanjiao Ying
- College of Automation, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China.,Institute of Biomedical Engineering and Instrument, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China
| | - Lei Zhu
- College of Automation, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China.,Institute of Biomedical Engineering and Instrument, Hangzhou Dianzi University, Hangzhou, Zhejiang, 310018, PR China
| |
Collapse
|
34
|
Martins WMBS, Li M, Sands K, Lenzi MH, Portal E, Mathias J, Dantas PP, Migliavacca R, Hunter JR, Medeiros EA, Gales AC, Toleman MA. Effective phage cocktail to combat the rising incidence of extensively drug-resistant Klebsiella pneumoniae sequence type 16. Emerg Microbes Infect 2022; 11:1015-1023. [PMID: 35259067 PMCID: PMC9004492 DOI: 10.1080/22221751.2022.2051752] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/06/2022] [Indexed: 11/18/2022]
Abstract
Bacteriophages are the most abundant organisms on Earth. As there are few effective treatment options against some pathogens, the interest in the bacteriophage control of multi-drug-resistant bacterial pathogens is escalating, especially for Klebsiella pneumoniae. This study aimed to develop a phage-based solution to the rising incidence of extensively drug-resistant clinical Klebsiella pneumoniae sequence type (ST16) infections starting from a set of phages recently characterized against this lineage. A phage-cocktail (Katrice-16) composed of eight lytic phages was characterized for potential use in humans. In vitro and in vivo broth inhibition and Galleria mellonella rescue assays were used to demonstrate the efficacy of this approach using a collection of 56 strains of K. pneumoniae ST16, with distinct genetic backgrounds that were collected from clinical infections from four disparate nations. Additionally, Katrice-16 anti-biofilm activity, synergism with meropenem, and activity in human body fluids were also assessed. Katrice-16 was highly active in vitro against our K. pneumoniae ST16 collection (AUC% median = 86.48%; Q1 = 83.8%; Q2 = 96.85%; Q3 = 98.85%). It additionally demonstrated excellent in vivo activity in G. mellonella rescue assays, even with larvae infected by isolates that exhibited moderate in vitro inhibition. We measured significant anti-biofilm activity over 12 h (p = .0113) and synergic activity with meropenem. In addition, we also demonstrate that Katrice-16 maintained high activity in human body fluids. Our results indicate that our cocktail will likely be an effective solution for human infections with this increasingly prevalent and often highly resistant bacterial clone.
Collapse
Affiliation(s)
- Willames M. B. S. Martins
- Department of Medical Microbiology, Division of Infection and Immunity, Cardiff University, Cardiff, UK
- Division of Infectious Diseases, Department of Internal Medicine, Escola Paulista de Medicina/Universidade Federal de São Paulo - UNIFESP, São Paulo, Brazil
| | - Mei Li
- Department of Medical Microbiology, Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Kirsty Sands
- Department of Medical Microbiology, Division of Infection and Immunity, Cardiff University, Cardiff, UK
- Department of Zoology, University of Oxford, Oxford, UK
| | - Michael H. Lenzi
- Division of Infectious Diseases, Department of Internal Medicine, Escola Paulista de Medicina/Universidade Federal de São Paulo - UNIFESP, São Paulo, Brazil
| | - Edward Portal
- Department of Medical Microbiology, Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Jordan Mathias
- Department of Medical Microbiology, Division of Infection and Immunity, Cardiff University, Cardiff, UK
| | - Priscila P. Dantas
- Division of Infection Control and Hospital Epidemiology, Hospital São Paulo, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Roberta Migliavacca
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, Unit of Microbiology and Clinical Microbiology, University of Pavia, Pavia, Italy
| | - James R. Hunter
- Division of Infectious Diseases, Department of Internal Medicine, Escola Paulista de Medicina/Universidade Federal de São Paulo - UNIFESP, São Paulo, Brazil
| | - Eduardo A. Medeiros
- Division of Infection Control and Hospital Epidemiology, Hospital São Paulo, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana C. Gales
- Division of Infectious Diseases, Department of Internal Medicine, Escola Paulista de Medicina/Universidade Federal de São Paulo - UNIFESP, São Paulo, Brazil
| | - Mark A. Toleman
- Department of Medical Microbiology, Division of Infection and Immunity, Cardiff University, Cardiff, UK
| |
Collapse
|
35
|
Alshahrani AM, Ibrahim ME, Aldossary AK, Alghamdi MA, Ahmed OB, Bin Abdulhak AA. Molecular Epidemiology of Carbapenem-Resistant K. pneumoniae Clinical Isolates from the Adult Patients with Comorbidities in a Tertiary Hospital, Southern Saudi Arabia. Antibiotics (Basel) 2022; 11:antibiotics11121697. [PMID: 36551354 PMCID: PMC9774885 DOI: 10.3390/antibiotics11121697] [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: 10/28/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Hospitalized patients are likely to have chronic illnesses and are at an increased risk of mortality due to infection caused by MDR bacteria. We aimed to identify carbapenem-resistant genes carrying Klebsiella pneumoniae (K. pneumoniae) isolates and their risk factors recovered from adult patients with comorbidities. A cross-sectional study was carried out between April 2021 and December 2021 at King Abdullah Hospital (KAH) in Bisha province, Saudi Arabia. Seventy-one multi-drug resistant K. pneumoniae recovered from clinical samples and screened for carbapenemase genes of blaOXA-48-like, blaNDM-1, blaKPC, blaVIM, and blaIMP. Of 71 MDR K. pneumoniae examined, 47 (66.2%) isolates harbored various carbapenemase genes. The most prevalent single resistance gene was blaOXA-48-like (62.5%; n = 25), and 33.3% of them were recovered from sputum isolates. The blaNDM-1 gene was detected in 12 (30.0%) isolates, and eight of them were recovered from urine (n = 4) and blood (n = 4). Two (5.0%) single blaKPC genes were recovered from the sputum (n = 1) and blood (n = 1) isolates. In contrast, no blaIMP- and blaVIM-carrying isolates were detected. The co-existence of two resistance genes between blaOXA-48-like and blaNDM-1 was found in six strains, whereas only one strain was found to be produced in the three genes of blaNDM-1, blaKPC, and blaOXA-48-like. There were statistically significant associations between the presence of carbapenem-gene-carrying K. pneumoniae and patients' gender (χ2(1) = 5.94, p = 0.015), intensive care unit admission (χ2(1) = 7.649, p = 0.002), and chronic obstructive pulmonary disease (χ2(1) = 4.851, p = 0.028). The study highlighted the existence of carbapenemase-producing K. pneumoniae, particularly blaOXA-48-like and blaNDM-1, in patients with comorbidities. Our findings emphasize the importance of the molecular characterization of resistance-determinant-carrying bacterial pathogens as a part of infection control and prevention in hospital settings.
Collapse
Affiliation(s)
- Abdullah M. Alshahrani
- Department of Family Medicine, College of Medicine, University of Bisha, Bisha 67614, Saudi Arabia
| | - Mutasim E. Ibrahim
- Department of Basic Medical Sciences (Microbiology Unit), College of Medicine, University of Bisha, Bisha 67614, Saudi Arabia
- Correspondence: or ; Tel.: +966-502656995
| | - Ahmed K. Aldossary
- Department of Family Medicine, College of Medicine, University of Bisha, Bisha 67614, Saudi Arabia
| | - Mushabab A. Alghamdi
- Department of Internal Medicine, College of Medicine, University of Bisha, Bisha 67614, Saudi Arabia
| | - Omar B. Ahmed
- Department of Environmental and Health Research, The Custodian of the Two Holy Mosques Institute for Hajj and Umrah Research, Umm Al-Qura University, Makkah 24382, Saudi Arabia
| | - Aref A. Bin Abdulhak
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52240, USA
| |
Collapse
|
36
|
A Comprehensive Genomic Analysis of the Emergent Klebsiella pneumoniae ST16 Lineage: Virulence, Antimicrobial Resistance and a Comparison with the Clinically Relevant ST11 Strain. Pathogens 2022; 11:pathogens11121394. [PMID: 36558729 PMCID: PMC9781218 DOI: 10.3390/pathogens11121394] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 11/24/2022] Open
Abstract
Klebsiella pneumoniae is considered an opportunistic pathogen frequently involved with healthcare-associated infections. The genome of K. pneumoniae is versatile, harbors diverse virulence factors and easily acquires and exchanges resistance plasmids, facilitating the emergence of new threatening clones. In the last years, ST16 has been described as an emergent, clinically relevant strain, increasingly associated with outbreaks, and carrying virulence factors (such as ICEKp, iuc, rmpADC/2) and a diversity of resistance genes. However, a far-reaching phylogenetic study of ST16, including geographically, clinically and temporally distributed isolates is not available. In this work, we analyzed all publicly available ST16 K. pneumoniae genomes in terms of virulence factors, including capsular lipopolysaccharide and polysaccharide diversity, plasmids and antimicrobial resistance genes. A core genome SNP analysis shows that less than 1% of studied sites were variant sites, with a median pairwise single nucleotide polymorphism difference of 87 SNPs. The number and diversity of antimicrobial resistance genes, but not of virulence-related genes, increased consistently in ST16 strains during the studied period. A genomic comparison between ST16 and the high-risk clone ST11 K. pneumoniae, showed great similarities in their capacity to acquire resistance and virulence markers, differing mostly in the great diversity of capsular lipopolysaccharide and polysaccharide types in ST11, in comparison with ST16. While virulence and antimicrobial resistance scores indicated that ST11 might still constitute a more difficult-to-manage strain, results presented here demonstrate the great potential of the ST16 clone becoming critical in public health.
Collapse
|
37
|
He Z, Xu W, Zhao H, Li W, Dai Y, Lu H, Zhao L, Zhang C, Li Y, Sun B. Epidemiological characteristics an outbreak of ST11 multidrug-resistant and hypervirulent Klebsiella pneumoniae in Anhui, China. Front Microbiol 2022; 13:996753. [PMID: 36212848 PMCID: PMC9537591 DOI: 10.3389/fmicb.2022.996753] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/22/2022] [Indexed: 11/29/2022] Open
Abstract
Klebsiella pneumoniae has become a primary threat to global health because of its virulence and resistance. In 2015, China reported multidrug-resistant (MDR) and hypervirulent K. pneumoniae (hvKp) isolates. The emergence of MDR-hvKp poses a significant threat to public health. We collected 76 MDR K. pneumoniae isolates from the same hospital, of which there were a total of six MDR-hvKp isolates. We performed multilocus sequence typing (MLST) and capsular typing, whole genome sequencing, comparative genome analysis, and phylogenetic analysis as well as phenotypic experiments, including growth curves, mucoviscosity assay, Galleria mellonella infection model, human whole blood survival, and human neutrophil bactericidal assay to further characterize the samples. We identified six large plasmids carrying extended spectrum β-lactamase (ESBL) genes or carbapenemase genes (blaCTX–M–65, blaKPC–2, blaSHV–12, blaSHV–158), 9 plasmids containing other drug resistance genes, and 7 hypervirulence plasmids carrying rmpA and rmpA2 in ST11 MDR-hvKp isolates. Some of these plasmids were identical, whereas others differed only by insertion elements. In addition, we identified a plasmid, p21080534_1, that carries hypervirulence genes (iucABCD, iutA, rmpA2), a carbapenemase gene (blaKPC–2), and an ESBL gene (blaSHV–12), as well as MDR-hvKp 21072329, which did not carry rmpA or rmpA2, but exhibited hypervirulence and hypermucoviscosity. ST11 MDR-hvKp derived from hypervirulence and multidrug resistance plasmids not only causes significant treatment difficulties, but also represents an unprecedented challenge to public health. Therefore, urgent measures are needed to limit further spread.
Collapse
Affiliation(s)
- Zhien He
- Department of Oncology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
- School of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Weifeng Xu
- Department of Oncology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
- School of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hang Zhao
- Department of Oncology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
- College of Life Science and Technology, Mudanjiang Normal University, Mudanjiang, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
- School of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yuanyuan Dai
- Department of Clinical Laboratory, Anhui Provincial Hospital of Anhui Medical University of China, Hefei, China
| | - Huaiwei Lu
- Department of Clinical Laboratory, Anhui Provincial Hospital of Anhui Medical University of China, Hefei, China
| | - Liping Zhao
- School of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Changfeng Zhang
- Clinical Laboratory Center, First Affiliated Hospital, Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Yujie Li
- Department of Oncology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
- School of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Yujie Li,
| | - Baolin Sun
- Department of Oncology, The First Affiliated Hospital, University of Science and Technology of China, Hefei, China
- School of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Baolin Sun,
| |
Collapse
|
38
|
Identification of the BolA Protein Reveals a Novel Virulence Factor in K. pneumoniae That Contributes to Survival in Host. Microbiol Spectr 2022; 10:e0037822. [PMID: 36121239 PMCID: PMC9603091 DOI: 10.1128/spectrum.00378-22] [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] [Indexed: 11/28/2022] Open
Abstract
BolA has been characterized as an important transcriptional regulator, which is induced in the stationary phase of growth and is often associated with bacterial virulence. This study was initiated to elucidate the role of the BolA in the virulence of K. pneumoniae. Using a mouse infection model, we revealed bolA mutant strain yielded significantly decreased bacterial loads in the liver, spleen, lung, and kidney, and failed to form liver abscesses. Gene deletion demonstrated that the bolA was required for siderophore production, biofilm formation, and adhesion to human colon cancer epithelial cells HCT116. Quantitative reverse transcriptase PCR (RT-qPCR) indicated that BolA could impact the expression of pulK, pulF, pulE, clpV, vgrG, entE, relA, and spoT genes on a genome-wide scale, which are related to type II secretion system (T2SS), type VI secretion system (T6SS), guanosine tetraphosphate (ppGpp), and siderophore synthesis and contribute to fitness in the host. Furthermore, the metabolome analysis showed that the deletion of the bolA gene led to decreased pools of five metabolites: biotin, spermine, cadaverine, guanosine, and flavin adenine dinucleotide, all of which are involved in pathways related to virulence and stress resistance. Taken together, we provided evidence that BolA was a significant virulence factor in the ability of K. pneumoniae to survive, and this was an important step in progress to an understanding of the pathways underlying bacterial virulence. IMPORTANCE BolA has been characterized as an important transcriptional regulator, which is induced in the stationary phase of growth and affects different pathways directly associated with bacterial virulence. Here, we unraveled the role of BolA in several phenotypes associated with the process of cell morphology, siderophore production, biofilm formation, cell adhesion, tissue colonization, and liver abscess. We also uncovered the importance of BolA for the success of K. pneumoniae infection and provided new clues to the pathogenesis strategies of this organism. This work constitutes a relevant step toward an understanding of the role of BolA protein as a master regulator and virulence factor. Therefore, this study is of great importance for understanding the pathways underlying K. pneumoniae virulence and may contribute to public health care applications.
Collapse
|
39
|
Abstract
Klebsiella pneumoniae (K. pneumoniae) is one of the most common pathogens causing nosocomial infection. A rapid, accurate, and convenient detection method is required for early diagnosis and directed therapy of K. pneumoniae infection. CRISPR-top (CRISPR-mediated testing in one pot) is a LAMP-CRISPR-based nucleic acid detection platform, which integrates target preamplification with CRISPR/Cas12b-based detection into a one-pot reaction mixture, performed at a constant temperature. In this study, we established the K. pneumoniae CRISPR-top assay to precisely identify K. pneumoniae at 56°C within 60 min. The reaction mixture with 0.53 μM (each) FIP and BIP, 0.27 μM LF, 0.13 μM (each) F3 and B3, and 2 μM ssDNA fluorescence probe was determined as the optimal reaction system of our assay. The limit of detection of this assay is 1 pg genomic DNA (equivalent to 160 K. pneumoniae cells and 1.6 × 105 CFU/mL for samples) per reaction, which is 10-fold more sensitive than LAMP. Up to 105 strains composed of K. pneumoniae clinical isolates and non-K. pneumoniae strains were correctly identified by our assay. A total of 58 sputum samples collected from patients with respiratory symptoms were used to evaluate the diagnostic performance of the K. pneumoniae CRISPR-top assay. As a result, the K. pneumoniae CRISPR-top assay yielded 100% (33/33) specificity and 96% (24/25) sensitivity, as well as a positive predictive value of 100% (24/24) and a negative predictive value of 97.1% (33/34), which were all higher than LAMP detection. In conclusion, the K. pneumoniae CRISPR-top assay developed in this study is a simple, rapid and ultra-specific method to detect K. pneumoniae. IMPORTANCEKlebsiella pneumoniae is a significant threat to global health. At present, the methods of K. pneumoniae detection are culture-based and instrument-dependent and are not suitable for rapid diagnostic. This study reports K. pneumoniae CRISPR-top assay, which can precisely identify K. pneumoniae using nucleic acids of pure cultures or clinical samples in one pot with one fluid-handling step. The K. pneumoniae CRISPR-top reaction can be completed within 60 min at a constant temperature, thus specific instruments are not required. Our results show that CRISPR-top assay yields enormous advantages compared with LAMP detection. The K. pneumoniae CRISPR-top assay can be a high-efficiency alternative tool for rapid and accurate diagnosis of K. pneumoniae infection, especially in resource-limited settings.
Collapse
|
40
|
Narimisa N, Goodarzi F, Bavari S. Prevalence of colistin resistance of Klebsiella pneumoniae isolates in Iran: a systematic review and meta-analysis. Ann Clin Microbiol Antimicrob 2022; 21:29. [PMID: 35765073 PMCID: PMC9241315 DOI: 10.1186/s12941-022-00520-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/07/2022] [Indexed: 11/11/2022] Open
Abstract
Objective Klebsiella pneumoniae is a gram-negative pathogen common cause of nosocomial infections. Colistin is a last resort antibiotic to treat infections caused by K. pneumoniae. In recent years, the resistance rate to colistin has increased in K. pneumoniae. This study evaluated the prevalence of colistin resistance of K. pneumoniae isolates in Iran using a systematic review and meta-analysis. Method A systematic search was performed for relevant articles until August 2021 in the following database: PubMed, Scopus, SID and Google Scholar. The pooled prevalence of colistin resistance in clinical K. pneumoniae isolates analyzed using Comprehensive Meta-Analysis Software (CMA). Results Finally, 19 articles with appropriate criteria were included in the meta-analysis. Our results showed 6.9% of the pooled prevalence of colistin resistance in clinical K. pneumoniae isolates in Iran. The results of subgroup analysis demonstrated increase resistance of colistin from 4.8%; (95% CI 1.5–13.9%) in 2013–2018 to 8.2%; (95% CI 3.4–18.6%), in 2019–2021. Also, the results of our study showed a strong association between the carbapenem producing K. pneumoniae and increased resistance to colistin. Conclusions This study showed a high prevalence of colistin resistance in K. pneumoniae isolates. It is recommended that regular evaluation be performed to control colistin resistance. Supplementary Information The online version contains supplementary material available at 10.1186/s12941-022-00520-8.
Collapse
Affiliation(s)
- Negar Narimisa
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Forough Goodarzi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Shirin Bavari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Erdem F, Díez-Aguilar M, Oksuz L, Kayacan C, Abulaila A, Oncul O, Morosini MI, Cantón R, Aktas Z. Time kill-assays of antibiotic combinations for multidrug resistant clinical isolates of OXA-48 carbapenemase producing Klebsiella pneumoniae. Acta Microbiol Immunol Hung 2022; 69:215-219. [PMID: 35895557 DOI: 10.1556/030.2022.01785] [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: 05/21/2022] [Accepted: 06/02/2022] [Indexed: 11/19/2022]
Abstract
Treatment of infections caused by OXA-48 carbapenemase producing multidrug-resistant isolates often necessitates combination therapy. In vitro effect of different antibiotic combinations against multidrug-resistant (MDR) Klebsiella pneumoniae isolates were evaluated in this study. Meropenem-tobramycin (MER+TOB), meropenem-ciprofloxacin (MER+CIP), colistin-meropenem (COL+MER), colistin-ciprofloxacin (COL+CIP) and colistin-tobramycin (COL+TOB) combinations were tested by time kill-assays. Each antibiotic alone and in combination at their Cmax values were tested against 4 clinical K. pneumoniae isolates at 1, 2, 4, 6, 8, 12 and 24 h. Effect of colistin and its associations were also assessed at 30 min. Bactericidal activity was defined as ≥3log10 CFU mL-1 decrease compared with initial inoculum. Synergy was defined as ≥2log10CFU mL-1 decrease by the combination compared with the most active single agent. Presence of bla OXA-48, bla NDM, bla VIM, bla IMP, bla KPC and bla CTX-M-1 genes was screened by PCR using specific primers. The bla OXA-48 gene was identified together with bla CTXM-1 group gene in all isolates. COL+MER demonstrated to be synergistic and bactericidal. MER+TOB showed synergistic and bactericidal effect on two strains although, regrowth was seen on other two strains at 24 h. MER+CIP exhibited indifferent effect on the strains. Combination therapy could be a potential alternative to treat MDR K. pneumoniae infections. This combination might prevent resistance development and secondary effects of colistin monotherapy. MER+TOB and MER+CIP might have an isolate-dependent effect, that may not always result in synergism.
Collapse
Affiliation(s)
- Fatma Erdem
- 1 Department of Medical Microbiology, Adana City Training and Research Hospital, Department of Medical Microbiology, Adana, Turkey
| | - María Díez-Aguilar
- 2 Servicio de Microbiología, Hospital Universitario La Princesa, Madrid, Spain
| | - Lutfiye Oksuz
- 7 Department of Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - Cigdem Kayacan
- 3 Department of Medical Microbiology, Faculty of Medicine, Istanbul Aydın University, Turkey
| | - Ayham Abulaila
- 4 Department of Clinical Microbiology, Istinye Faculty of Medicine, Istinye University, Istanbul, Turkey
| | - Oral Oncul
- 5 Department of Infectious Diseases and Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Turkey
| | - María Isabel Morosini
- 6 Servicio de Microbiología. Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Rafael Cantón
- 6 Servicio de Microbiología. Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Zerrin Aktas
- 7 Department of Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Turkey
| |
Collapse
|
42
|
Gorrie CL, Mirčeta M, Wick RR, Judd LM, Lam MMC, Gomi R, Abbott IJ, Thomson NR, Strugnell RA, Pratt NF, Garlick JS, Watson KM, Hunter PC, Pilcher DV, McGloughlin SA, Spelman DW, Wyres KL, Jenney AWJ, Holt KE. Genomic dissection of Klebsiella pneumoniae infections in hospital patients reveals insights into an opportunistic pathogen. Nat Commun 2022; 13:3017. [PMID: 35641522 PMCID: PMC9156735 DOI: 10.1038/s41467-022-30717-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/04/2022] [Indexed: 12/11/2022] Open
Abstract
Klebsiella pneumoniae is a major cause of opportunistic healthcare-associated infections, which are increasingly complicated by the presence of extended-spectrum beta-lactamases (ESBLs) and carbapenem resistance. We conducted a year-long prospective surveillance study of K. pneumoniae clinical isolates in hospital patients. Whole-genome sequence (WGS) data reveals a diverse pathogen population, including other species within the K. pneumoniae species complex (18%). Several infections were caused by K. variicola/K. pneumoniae hybrids, one of which shows evidence of nosocomial transmission. A wide range of antimicrobial resistance (AMR) phenotypes are observed, and diverse genetic mechanisms identified (mainly plasmid-borne genes). ESBLs are correlated with presence of other acquired AMR genes (median n = 10). Bacterial genomic features associated with nosocomial onset are ESBLs (OR 2.34, p = 0.015) and rhamnose-positive capsules (OR 3.12, p < 0.001). Virulence plasmid-encoded features (aerobactin, hypermucoidy) are observed at low-prevalence (<3%), mostly in community-onset cases. WGS-confirmed nosocomial transmission is implicated in just 10% of cases, but strongly associated with ESBLs (OR 21, p < 1 × 10-11). We estimate 28% risk of onward nosocomial transmission for ESBL-positive strains vs 1.7% for ESBL-negative strains. These data indicate that K. pneumoniae infections in hospitalised patients are due largely to opportunistic infections with diverse strains, with an additional burden from nosocomially-transmitted AMR strains and community-acquired hypervirulent strains.
Collapse
Affiliation(s)
- Claire L Gorrie
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Vic, Australia.
| | - Mirjana Mirčeta
- Microbiology Unit, Alfred Pathology Service, The Alfred Hospital, Melbourne, Vic, Australia
| | - Ryan R Wick
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Vic, Australia
| | - Louise M Judd
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Vic, Australia
- Doherty Applied Microbial Genomics (DAMG), Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Vic, Australia
| | - Margaret M C Lam
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Vic, Australia
| | - Ryota Gomi
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Vic, Australia
- Department of Environmental Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Iain J Abbott
- Microbiology Unit, Alfred Pathology Service, The Alfred Hospital, Melbourne, Vic, Australia
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Vic, Australia
| | - Nicholas R Thomson
- Wellcome Sanger Institute, Hinxton, Cambs, UK
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK
| | - Richard A Strugnell
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Vic, Australia
| | - Nigel F Pratt
- Infectious Diseases Clinical Research Unit, The Alfred Hospital, Melbourne, Vic, Australia
| | - Jill S Garlick
- Infectious Diseases Clinical Research Unit, The Alfred Hospital, Melbourne, Vic, Australia
| | - Kerrie M Watson
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Vic, Australia
| | - Peter C Hunter
- Aged Care, Caulfield Hospital, Alfred Health, Melbourne, Vic, Australia
| | - David V Pilcher
- Intensive Care Unit, The Alfred Hospital, Melbourne, Vic, Australia
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventative Medicine, Monash University, Melbourne, Vic, Australia
| | - Steve A McGloughlin
- Intensive Care Unit, The Alfred Hospital, Melbourne, Vic, Australia
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventative Medicine, Monash University, Melbourne, Vic, Australia
| | - Denis W Spelman
- Microbiology Unit, Alfred Pathology Service, The Alfred Hospital, Melbourne, Vic, Australia
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Vic, Australia
| | - Kelly L Wyres
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Vic, Australia
| | - Adam W J Jenney
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Vic, Australia
- Microbiology Unit, Alfred Pathology Service, The Alfred Hospital, Melbourne, Vic, Australia
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Vic, Australia
| | - Kathryn E Holt
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Vic, Australia.
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK.
| |
Collapse
|
43
|
Le MNT, Kayama S, Wyres KL, Yu L, Hisatsune J, Suzuki M, Yahara K, Terachi T, Sawa K, Takahashi S, Okuhara T, Kohama K, Holt KE, Mizutani T, Ohge H, Sugai M. Genomic epidemiology and temperature dependency of hypermucoviscous Klebsiella pneumoniae in Japan. Microb Genom 2022; 8. [PMID: 35622495 PMCID: PMC9465067 DOI: 10.1099/mgen.0.000827] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Klebsiella pneumoniae (Kp) has emerged as a global life-threatening pathogen owing to its multidrug resistance and hypervirulence phenotype. Several fatal outbreaks of carbapenem-resistant hypervirulent Kp have been reported recently. Hypermucoviscosity (HMV) is a phenotype commonly associated with hypervirulence of Kp, which is usually regulated by rmpA or rmpA2 (regulators of the mucoid phenotype). Here, we found that temperature was important in the HMV phenotype of Kp, and the impact of temperature on HMV was not uniform among strains. We investigated the HMV phenotype at 37 °C and room temperature (20–25 °C) in 170 clinically isolated hypermucoviscous Kp strains in Japan and analysed the association between the HMV phenotype, virulence genes and antimicrobial resistance (AMR) genes. String length distribution at different temperatures was correlated with the genomic population of Kp. The strains carrying rmpA/rmpA2 frequently showed the HMV phenotype at 37 °C, while the strains negative for these genes tended to show the HMV phenotype at room temperature. Hypervirulent Kp clusters carrying rmpA/rmpA2 without extended-spectrum beta-lactamases (ESBL)/carbapenemases produced higher string lengths at 37 °C than at room temperature, and were mostly isolated from the respiratory tract. Other HMV strains showed distinct characteristics of not carrying rmpA/rmpA2 but were positive for ESBL/carbapenemases, with a higher string length at room temperature than at 37 °C, and were frequently isolated from bloodstream infections. In total, 21 (13.5 %) HMV isolates carried ESBL and carbapenemases, among which five isolates were carbapenem-resistant hypervirulent Kp with a pLVPK-like plasmid (an epidemic virulence plasmid) and a pKPI-6-like plasmid (an epidemic blaIMP-6-bearing plasmid in Japan), suggesting the convergence of worldwide hypervirulence and epidemic AMR in Japan.
Collapse
Affiliation(s)
- Mi Nguyen-Tra Le
- Department of Antimicrobial Resistance, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan.,Project Research Center for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan.,Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Shizuo Kayama
- Department of Antimicrobial Resistance, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan.,Project Research Center for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan.,Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Japan
| | - Kelly L Wyres
- Department of Infectious Diseases, Monash University, Melboune, Victoria, Australia
| | - Liansheng Yu
- Department of Antimicrobial Resistance, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan.,Project Research Center for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan.,Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Japan
| | - Junzo Hisatsune
- Department of Antimicrobial Resistance, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan.,Project Research Center for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan.,Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Japan
| | - Masato Suzuki
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Japan
| | - Koji Yahara
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Japan
| | - Tsuneko Terachi
- Department of Clinical Laboratory, Osaka Police Hospital, Osaka, Japan
| | - Kana Sawa
- Department of Clinical Laboratory, Osaka Police Hospital, Osaka, Japan
| | | | | | - Kunihiko Kohama
- Department of Clinical Laboratory, Chugoku Rosai Hospital, Hiroshima, Japan
| | - Kathryn E Holt
- Department of Infectious Diseases, Monash University, Melboune, Victoria, Australia.,London School of Hygiene and Tropical Medicine, London, UK
| | - Tetsu Mizutani
- Department of Clinical Laboratory, Osaka Police Hospital, Osaka, Japan
| | - Hiroki Ohge
- Department of Infectious Diseases, Hiroshima University Hospital, Hiroshima, Japan.,Project Research Center for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan
| | - Motoyuki Sugai
- Department of Antimicrobial Resistance, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan.,Project Research Center for Nosocomial Infectious Diseases, Hiroshima University, Hiroshima, Japan.,Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Japan
| |
Collapse
|
44
|
Hawkey J, Cottingham H, Tokolyi A, Wick RR, Judd LM, Cerdeira L, de Oliveira Garcia D, Wyres KL, Holt KE. Linear plasmids in Klebsiella and other Enterobacteriaceae. Microb Genom 2022; 8. [PMID: 35416146 PMCID: PMC9453081 DOI: 10.1099/mgen.0.000807] [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] [Indexed: 11/20/2022] Open
Abstract
Linear plasmids are extrachromosomal DNA elements that have been found in a small number of bacterial species. To date, the only linear plasmids described in the family Enterobacteriaceae belong to Salmonella, first found in Salmonella enterica Typhi. Here, we describe a collection of 12 isolates of the Klebsiella pneumoniae species complex in which we identified linear plasmids. Screening of assembly graphs assembled from public read sets identified linear plasmid structures in a further 13 K. pneumoniae species complex genomes. We used these 25 linear plasmid sequences to query all bacterial genome assemblies in the National Center for Biotechnology Information database, and discovered an additional 61 linear plasmid sequences in a variety of Enterobacteriaceae species. Gene content analysis divided these plasmids into five distinct phylogroups, with very few genes shared across more than two phylogroups. The majority of linear plasmid-encoded genes are of unknown function; however, each phylogroup carried its own unique toxin–antitoxin system and genes with homology to those encoding the ParAB plasmid stability system. Passage in vitro of the 12 linear plasmid-carrying Klebsiella isolates in our collection (which include representatives of all five phylogroups) indicated that these linear plasmids can be stably maintained, and our data suggest they can transmit between K. pneumoniae strains (including members of globally disseminated multidrug-resistant clones) and also between diverse Enterobacteriaceae species. The linear plasmid sequences, and representative isolates harbouring them, are made available as a resource to facilitate future studies on the evolution and function of these novel plasmids.
Collapse
Affiliation(s)
- Jane Hawkey
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Hugh Cottingham
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Alex Tokolyi
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Ryan R Wick
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Louise M Judd
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | | | | | - Kelly L Wyres
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Kathryn E Holt
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia.,Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| |
Collapse
|
45
|
Reduced Virulence and Enhanced Host Adaption during Antibiotics Therapy: a Story of a Within-Host Carbapenem-Resistant Klebsiella pneumoniae Sequence Type 11 Evolution in a Patient with a Serious Scrotal Abscess. mSystems 2022; 7:e0134221. [PMID: 35196132 PMCID: PMC9040587 DOI: 10.1128/msystems.01342-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Carbapenem-resistant Klebsiella pneumoniae (CRKP) has disseminated globally and threatened human life. The sequence type (ST) 11 CRKP is a dominant clone in Asia, but how this clone evolves in vivo then adapts to the host and facilitates dissemination remains largely unknown. Here, the genomic dynamics of 4 ST11-CRKP isolates, which were sequentially collected from the urine of a patient with initial serious scrotal abscess and finally recovered without effective medication, were analyzed. Genomic differences were identified and their implications for pathogenesis and host adaptation were investigated. The related transcriptional pathways were further explored by RNA-Seq. Genomic analysis identified 4 to 24 mutations, among which 94% to 100% of them were synonymous or intergenic mutations. During 47 days of antibiotics therapy, CRKP underwent adaptive evolution, including tigecycline resistance and virulence attenuation. Tigecycline resistance was caused by a deletion within the ramR ribosomal binding site, which has been described by us previously. On the other hand, mutations associated with two genes, acyltransferase (act) and ompK26, resulted in the attenuation phenotype of ST11-CRKP. act deficiency reduced the capsular polysaccharide (CPS) production, enhanced biofilm formation, weakened capsular protection, and decreased induction of proinflammatory cytokines. Further RNA-Seq analysis revealed that act influenced the expression of ldhA, bglX, mtnK, and metE which likely participate in capsular synthesis and biofilm formation. ompK26 affected the virulence by its overexpression caused by the deletion of the upstream repressor binding site. This study presents a within-host adaption of ST11-CRKP and suggests an important role of CPS in the adaptive evolution of virulence and persistence of CRKP. IMPORTANCE Carbapenem-resistant Klebsiella pneumoniae (CRKP) has disseminated worldwide and can cause life-threatening infections, including pneumonia, bloodstream infections, urinary tract infections, intraabdominal infection, liver abscess, and meningitis. CRKP infection is the leading cause of high mortality in hospitals. The sequence type (ST) 11 CRKP is a dominant clone and accounts for 60% of CRKP infections in China. Recently, the ST11-CRKP with high transmissibility is increasingly identified. Understanding how this clone has evolved is crucial for developing strategies to control its further dissemination. The significance of our research is the identification of the in vivo genomic dynamics of ST11-CRKP and the genetic basis for ST11-CRKP that facilitate persistence and dissemination. Furthermore, our study also highlights the importance of monitoring the within-host evolution of pathogens during the treatment and developing interventions to minimize the potential impact of host adaptation on human health.
Collapse
|
46
|
Hong Nguyen M, Shields RK, Chen L, William Pasculle A, Hao B, Cheng S, Sun J, Kline EG, Kreiswirth BN, Clancy CJ. Molecular Epidemiology, Natural History, and Long-Term Outcomes of Multidrug-Resistant Enterobacterales Colonization and Infections Among Solid Organ Transplant Recipients. Clin Infect Dis 2022; 74:395-406. [PMID: 33970222 PMCID: PMC8834656 DOI: 10.1093/cid/ciab427] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Multidrug-resistant Enterobacterales (MDR-E), including carbapenem-resistant and third-generation cephalosporin-resistant Enterobacterales (CRE, CefR-E), are major pathogens following solid organ transplantation (SOT). METHODS We prospectively studied patients who underwent lung, liver, and small bowel transplant from February 2015 through March 2017. Weekly perirectal swabs (up to 100 days post-transplant) were cultured for MDR-E. Whole-genome sequencing (WGS) was performed on gastrointestinal (GI) tract-colonizing and disease-causing isolates. RESULTS Twenty-five percent (40 of 162) of patients were MDR-E GI-colonized. Klebsiella pneumoniae was the most common CRE and CefR-E. Klebsiella pneumoniae carbapenemases and CTX-M were leading causes of CR and CefR, respectively. Thirty-five percent of GI colonizers developed MDR-E infection vs 2% of noncolonizers (P < .0001). The attack rate was higher among CRE colonizers than CefR-E colonizers (53% vs 21%, P = .049). GI colonization and high body mass index were independent risk factors for MDR-E infection (P ≤ .004). Thirty-day mortality among infected patients was 6%. However, 44% of survivors developed recurrent infections; 43% of recurrences were late (285 days to 3.9 years after the initial infection). Long-term survival (median, 4.3 years post-transplant) did not differ significantly between MDR-E-infected and MDR-E-noninfected patients (71% vs 77%, P = .56). WGS phylogenetic analyses revealed that infections were caused by GI-colonizing strains and suggested unrecognized transmission of novel clonal group-258 sublineage CR-K. pneumoniae and horizontal transfer of resistance genes. CONCLUSIONS MDR-E GI colonization was common following SOT and predisposed patients to infections by colonizing strains. MDR-E infections were associated with low short- and long-term mortality, but recurrences were frequent and often occurred years after initial infections. Findings provide support for MDR-E surveillance in our SOT program.
Collapse
Affiliation(s)
- M Hong Nguyen
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- XDR Pathogen Laboratory, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Division of Infectious Diseases, Transplant Infectious Diseases, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ryan K Shields
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- XDR Pathogen Laboratory, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Liang Chen
- Hackensack-Meridian Health Center for Discovery and Innovation, Nutley, New Jersey, USA
| | - A William Pasculle
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Binghua Hao
- XDR Pathogen Laboratory, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Shaoji Cheng
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jonathan Sun
- XDR Pathogen Laboratory, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ellen G Kline
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Barry N Kreiswirth
- Division of Infectious Diseases, Transplant Infectious Diseases, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Cornelius J Clancy
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- XDR Pathogen Laboratory, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- VA Pittsburgh Healthcare System, Department of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
47
|
Hou J, Mao D, Zhang Y, Huang R, Li L, Wang X, Luo Y. Long-term spatiotemporal variation of antimicrobial resistance genes within the Serratia marcescens population and transmission of S. marcescens revealed by public whole-genome datasets. JOURNAL OF HAZARDOUS MATERIALS 2022; 423:127220. [PMID: 34844350 DOI: 10.1016/j.jhazmat.2021.127220] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/05/2021] [Accepted: 09/10/2021] [Indexed: 06/13/2023]
Abstract
The development of antimicrobial resistance (AMR) is accelerated by the selective pressure exerted by the widespread use of antimicrobial drugs, posing an increasing danger to public health. However, long-term spatiotemporal variation in AMR genes in microorganisms, particularly in bacterial pathogens in response to antibiotic consumption, is not fully understood. Here, we used the NCBI RefSeq database to collect 478 whole-genome sequences for Serratia marcescens ranging from 1961 up to 2019, to document global long-term AMR trends in S. marcescens populations. In total, 100 AMR gene subtypes (16 AMR gene types) were detected in the genomes of S. marcescens populations. We identified 3 core resistance genes in S. marcescens genomes, and a high diversity of AMR genes was observed in S. marcescens genomes after corresponding antibiotics were discovered and introduced into clinical practice, suggesting the adaptation of S. marcescens populations to challenges with therapeutic antibiotics. Our findings indicate spatiotemporal variation of AMR genes in S. marcescens populations in relation to antibiotic consumption and suggest the potential transmission of S. marcescens isolates harboring AMR genes among countries and between the environment and the clinic, representing a public health threat that necessitates international solidarity to overcome.
Collapse
Affiliation(s)
- Jie Hou
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin 300071, China
| | - Daqing Mao
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Yulin Zhang
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin 300071, China
| | - Ruiyang Huang
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin 300071, China
| | - Linyun Li
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin 300071, China
| | - Xiaolong Wang
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin 300071, China
| | - Yi Luo
- College of Environmental Science and Engineering, Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, Nankai University, Tianjin 300071, China; State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210093, China.
| |
Collapse
|
48
|
Fostervold A, Hetland MAK, Bakksjø R, Bernhoff E, Holt KE, Samuelsen Ø, Simonsen GS, Sundsfjord A, Wyres KL, Löhr IH. A nationwide genomic study of clinical Klebsiella pneumoniae in Norway 2001-15: introduction and spread of ESBLs facilitated by clonal groups CG15 and CG307. J Antimicrob Chemother 2021; 77:665-674. [PMID: 34935048 PMCID: PMC8865009 DOI: 10.1093/jac/dkab463] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/12/2021] [Indexed: 01/21/2023] Open
Abstract
Objectives To use the nationwide Norwegian surveillance programme on resistant microbes in humans (NORM) to address longitudinal changes in the population structure of Klebsiella pneumoniae isolates from 2001–15, focusing on the emergence and dissemination of ESBL-producing K. pneumoniae in Norway. Methods Among blood (n = 6124) and urinary tract (n = 5496) surveillance isolates from 2001–15, we used Illumina technology to whole genome sequence 201 ESBL-producing isolates from blood (n = 130) and urine (n = 71), and 667 non-ESBL isolates from blood. Complete genomes for four isolates were resolved with Oxford Nanopore sequencing. Results In a highly diverse collection, Klebsiella variicola ssp. variicola caused 24.5% of Klebsiella pneumoniae species complex (KpSC) bacteraemias. ESBL production was limited to K. pneumoniae sensu stricto (98.5%). A diverse ESBL population of 57 clonal groups (CGs) were dominated by MDR CG307 (17%), CG15 (12%), CG70 (6%), CG258 (5%) and CG45 (5%) carrying blaCTX-M-15. Yersiniabactin was significantly more common in ESBL-positive (37.8%) compared with non-ESBL K. pneumoniae sensu stricto isolates (12.7%), indicating convergence of virulence and resistance determinants. Moreover, we found a significantly lower prevalence of yersiniabactin (3.0%, 37.8% and 17.3%), IncFIB (58.7%, 87.9% and 79.4%) and IncFII plasmid replicons (40.5%, 82.8% and 54.2%) in K. variicola ssp. variicola compared with ESBL- and non-ESBL K. pneumoniae sensu stricto isolates, respectively. Conclusions The increase in Norwegian ESBL-producing KpSC during 2010–15 was driven by CG307 and CG15 carrying blaCTX-M-15. K. variicola ssp. variicola was a frequent cause of invasive KpSC infection, but rarely carried ESBLs.
Collapse
Affiliation(s)
- Aasmund Fostervold
- Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway.,Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Marit A K Hetland
- Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway.,Department of Biological Sciences, Faculty of Mathematics and Natural Sciences, University of Bergen, Bergen, Norway
| | - Ragna Bakksjø
- Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway
| | - Eva Bernhoff
- Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway
| | - Kathryn E Holt
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Australia.,Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Ørjan Samuelsen
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, University Hospital of North Norway, Tromsø, Norway.,Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Gunnar Skov Simonsen
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Microbiology and Infection Control, University Hospital of North-Norway, Tromsø, Norway
| | - Arnfinn Sundsfjord
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, University Hospital of North Norway, Tromsø, Norway.,Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Kelly L Wyres
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Australia
| | - Iren Høyland Löhr
- Department of Medical Microbiology, Stavanger University Hospital, Stavanger, Norway.,Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | | |
Collapse
|
49
|
Cornick J, Musicha P, Peno C, Seager E, Iroh Tam PY, Bilima S, Bennett A, Kennedy N, Feasey N, Heinz E, Cain AK. Genomic investigation of a suspected Klebsiella pneumoniae outbreak in a neonatal care unit in sub-Saharan Africa. Microb Genom 2021; 7. [PMID: 34793293 PMCID: PMC8743538 DOI: 10.1099/mgen.0.000703] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A special-care neonatal unit from a large public hospital in Malawi was noted as having more frequent, difficult-to-treat infections, and a suspected outbreak of multi-drug-resistant Klebsiella pneumoniae was investigated using genomic characterisation. All K. pneumoniae bloodstream infections (BSIs) from patients in the neonatal ward (n=62), and a subset of K. pneumoniae BSI isolates (n=38) from other paediatric wards in the hospital, collected over a 4 year period were studied. After whole genome sequencing, the strain sequence types (STs), plasmid types, virulence and resistance genes were identified. One ST340 clone, part of clonal complex 258 (CC258) and an ST that drives hospital outbreaks worldwide, harbouring numerous resistance genes and plasmids, was implicated as the likely cause of the outbreak. This study contributes molecular information necessary for tracking and characterizing this important hospital pathogen in sub-Saharan Africa.
Collapse
Affiliation(s)
- Jennifer Cornick
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi.,Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Patrick Musicha
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi.,Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Chikondi Peno
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi.,Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ezgi Seager
- Birmingham Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Bordesley Green East, Birmingham, B9 5SS, UK
| | - Pui-Ying Iroh Tam
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi.,Liverpool School of Tropical Medicine, Liverpool, UK
| | - Sithembile Bilima
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Aisleen Bennett
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi.,Institute for Infection and Immunity, St George's, University of London, London, UK
| | - Neil Kennedy
- Centre for Medical Education, Queen's University Belfast, Belfast, Ireland.,Department of Paediatrics, College of Medicine, University of Malawi, Zomba, Malawi
| | - Nicholas Feasey
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi.,Liverpool School of Tropical Medicine, Liverpool, UK
| | - Eva Heinz
- Pathogen Genomics, Wellcome Sanger Institute, Cambridge, UK.,Liverpool School of Tropical Medicine, Liverpool, UK
| | - Amy K Cain
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi.,Pathogen Genomics, Wellcome Sanger Institute, Cambridge, UK.,Department of Molecular Sciences, Macquarie University, Sydney, Australia.,Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
50
|
Han X, Shi Q, Mao Y, Quan J, Zhang P, Lan P, Jiang Y, Zhao D, Wu X, Hua X, Yu Y. Emergence of Ceftazidime/Avibactam and Tigecycline Resistance in Carbapenem-Resistant Klebsiella pneumoniae Due to In-Host Microevolution. Front Cell Infect Microbiol 2021; 11:757470. [PMID: 34760723 PMCID: PMC8573091 DOI: 10.3389/fcimb.2021.757470] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/08/2021] [Indexed: 11/19/2022] Open
Abstract
Klebsiella pneumoniae can cause both hospital- and community-acquired clinical infections. Last-line antibiotics against carbapenem-resistant K. pneumoniae (CRKP), such as ceftazidime/avibactam (CZA) and tigecycline (TGC), remain limited as treatment choices. This study aimed to investigate the mechanisms by which CRKP acquires CZA and TGC resistance in vivo under β-lactam antibiotic and TGC exposure. Three CRKP strains (XDX16, XDX31 and XDX51) were consecutively isolated from an inpatient with a urinary tract infection in two months. PFGE and MLST showed that these strains were closely related and belonged to sequence type (ST) 4496, which is a novel ST closely related to ST11. Compared to XDX16 and XDX31, XDX51 developed CZA and TGC resistance. Sequencing showed that double copies of blaKPC-2 were located on a 108 kb IncFII plasmid, increasing blaKPC-2 expression in XDX51. In addition, ramR was interrupted by Insertion sequence (IS) Kpn14 in XDX51, with this strain exhibiting upregulation of ramA, acrA and acrB expression compared with XDX16 and XDX31. Furthermore, LPS analysis suggested that the O-antigen in XDX51 was defective due to ISKpn26 insertion in the rhamnosyl transferase gene wbbL, which slightly reduced TGC susceptibility. In brief, CZA resistance was caused mainly by blaKPC-2 duplication, and TGC resistance was caused by ramR inactivation with additional LPS changes due to IS element insertion in wbbL. Notably, CRKP developed TGC and CZA resistance within one month under TGC and β-lactam treatment without exposure to CZA. The CRKP clone ST4496 has the ability to evolve CZA and TGC resistance rapidly, posing a potential threat to inpatients during antibiotic treatment.
Collapse
Affiliation(s)
- Xinhong Han
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiucheng Shi
- Department of Clinical Laboratory, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yihan Mao
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Quan
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ping Zhang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Lan
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Jiang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongdong Zhao
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xueqing Wu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoting Hua
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunsong Yu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|