1
|
Shen D, Seco BMS, Teixeira Alves LG, Yao L, Bräutigam M, Opitz B, Witzenrath M, Fries BC, Seeberger PH. Semisynthetic Glycoconjugate Vaccine Lead against Klebsiella pneumoniae Serotype O2afg Induces Functional Antibodies and Reduces the Burden of Acute Pneumonia. J Am Chem Soc 2024; 146:35356-35366. [PMID: 39666976 DOI: 10.1021/jacs.4c13972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Carbapenem-resistant Klebsiella pneumoniae (CR-Kp) bacteria are a serious global health concern due to their drug-resistance to nearly all available antibiotics, fast spread, and high mortality rate. O2afg is a major CR-Kp serotype in the sequence type 258 group (KPST258) that is weakly immunogenic in humans. Here, we describe the creation and evaluation of semisynthetic O2afg glycoconjugate vaccine leads containing one and two repeating units of the polysaccharide epitope that covers the surface of the bacteria conjugated to the carrier protein CRM197. The semisynthetic glycoconjugate containing two repeating units induced functional IgG antibodies in rabbits with opsonophagocytic killing activity and enhanced complement activation and complement-mediated killing of CR-Kp. Passive immunization reduced the burden of acute pneumonia in mice and may represent an alternative to antimicrobial therapy. The semisynthetic glycoconjugate vaccine lead against CR-Kp expressing O2afg antigen is awaiting preclinical development.
Collapse
Affiliation(s)
- Dacheng Shen
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Bruna M S Seco
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Luiz Gustavo Teixeira Alves
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charite-Universitätsmedizin Berlin; 10117 Berlin, Germany
| | - Ling Yao
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Maria Bräutigam
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Bastian Opitz
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charite-Universitätsmedizin Berlin; 10117 Berlin, Germany
- German Center for Lung Research (DZL), 12203 Berlin, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charite-Universitätsmedizin Berlin; 10117 Berlin, Germany
- German Center for Lung Research (DZL), 12203 Berlin, Germany
| | - Bettina C Fries
- Department of Medicine, Infectious Disease Division, Stony Brook University; Stony Brook, New York 11794, United States
- Veteran's Administration Medical Center, Northport, New York 11768, United States
| | - Peter H Seeberger
- Department of Bimolecular System, Max Planck Institute of Colloids and Interfaces; 14476 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| |
Collapse
|
2
|
Kawser Z, Sridhar S, Kar S, Habib T, Mukta SA, Azad K, Hasan N, Kulsum U, Siddik AB, Rahman S, Tanni NN, Nesa M, Earl AM, Worby CJ, Turbett SE, Shamsuzzaman SM, Harris JB, Qadri F, LaRocque RC. Clinical and genomic characterization of Klebsiella pneumoniae infections in Dhaka, Bangladesh. J Glob Antimicrob Resist 2024:S2213-7165(24)00483-1. [PMID: 39725322 DOI: 10.1016/j.jgar.2024.12.016] [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: 04/03/2024] [Revised: 11/05/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Klebsiella pneumoniae (Kpn), a WHO priority pathogen with high rates of antimicrobial resistance (AMR), has emerged as a leading cause of hospital acquired pneumonia and neonatal sepsis. OBJECTIVE We aimed to define the clinical characteristics of a cohort of patients with Kpn infection in Dhaka, Bangladesh and to perform phenotypic and genetic characterization of the associated isolates. METHODS We retrospectively extracted clinical data about patients at Dhaka Medical College Hospital from whom Klebsiella spp was isolated from a clinical specimen collected between February and September 2022. We used standard microbiologic techniques to evaluate AMR and whole-genome sequencing (WGS) to assess dominant lineages, common capsular (K) and O-polysaccharide (O) antigen types, and AMR and virulence genes. RESULTS Ninety-eight patients were included, with diagnoses of pneumonia (38/98, 39%), wound infection (29/98, 31%), urinary tract infection (29/98, 31%) and bacteremia (2/98, 2%). We tested isolates for susceptibility to eight classes of antibiotics. Of the 98 isolates, 41% were multidrug resistant (MDR), 15% were extensively drug resistant (XDR), and 16% were pan-drug resistant (PDR). Three isolates (3%) were resistant to polymyxin B. Outcome data were available for 46 patients; 4 patients (8%) died from infections caused by PDR (n=2), XDR (n=1), and MDR isolates (n=1). WGS revealed a high degree of genomic diversity, with multiple sequence types (STs), O-types and K-types represented; ST16:K81:OL101 and ST43:K30:O1 were the most prevalent. CONCLUSION Our findings suggest alarming levels of AMR among Kpn isolates in Bangladesh and a critical need for improved treatment modalities and vaccine development.
Collapse
Affiliation(s)
- Zannat Kawser
- institute for developing Science and Health initiatives, Dhaka, Bangladesh
| | - Sushmita Sridhar
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA USA
| | - Sanchita Kar
- institute for developing Science and Health initiatives, Dhaka, Bangladesh
| | - Tanbir Habib
- institute for developing Science and Health initiatives, Dhaka, Bangladesh
| | | | - Kasrina Azad
- institute for developing Science and Health initiatives, Dhaka, Bangladesh
| | - Neyamul Hasan
- institute for developing Science and Health initiatives, Dhaka, Bangladesh
| | - Umme Kulsum
- institute for developing Science and Health initiatives, Dhaka, Bangladesh
| | - Abu Bakar Siddik
- institute for developing Science and Health initiatives, Dhaka, Bangladesh
| | - Saikt Rahman
- institute for developing Science and Health initiatives, Dhaka, Bangladesh
| | | | - Maherun Nesa
- Dhaka Medical College and Hospital, Dhaka, Bangladesh
| | - Ashlee M Earl
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Colin J Worby
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Sarah E Turbett
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA USA; Harvard Medical School, Boston, MA USA; Department of Pathology, Massachusetts General Hospital, Boston, MA USA
| | | | - Jason B Harris
- Harvard Medical School, Boston, MA USA; Massachusetts General Hospital for Children, Boston, MA USA
| | - Firdausi Qadri
- institute for developing Science and Health initiatives, Dhaka, Bangladesh; Institutional Center for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Regina C LaRocque
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA USA; Harvard Medical School, Boston, MA USA.
| |
Collapse
|
3
|
Cheetham MJ, Huo Y, Stroyakovski M, Cheng L, Wan D, Dell A, Santini JM. Specificity and diversity of Klebsiella pneumoniae phage-encoded capsule depolymerases. Essays Biochem 2024; 68:661-677. [PMID: 39668555 DOI: 10.1042/ebc20240015] [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: 08/30/2024] [Revised: 11/26/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Klebsiella pneumoniae is an opportunistic pathogen with significant clinical relevance. K. pneumoniae-targeting bacteriophages encode specific polysaccharide depolymerases with the ability to selectively degrade the highly varied protective capsules, allowing for access to the bacterial cell wall. Bacteriophage depolymerases have been proposed as novel antimicrobials to combat the rise of multidrug-resistant K. pneumoniae strains. These enzymes display extraordinary diversity, and are key determinants of phage host range, however with limited data available our current knowledge of their mechanisms and ability to predict their efficacy is limited. Insight into the resolved structures of Klebsiella-specific capsule depolymerases reveals varied catalytic mechanisms, with the intra-chain cleavage mechanism providing opportunities for recombinant protein engineering. A detailed comparison of the 58 characterised depolymerases hints at structural and mechanistic patterns, such as the conservation of key domains for substrate recognition and phage tethering, as well as diversity within groups of depolymerases that target the same substrate. Another way to understand depolymerase specificity is by analyzing the targeted capsule structures, as these may share similarities recognizable by bacteriophage depolymerases, leading to broader substrate specificities. Although we have only begun to explore the complexity of Klebsiella capsule depolymerases, further research is essential to thoroughly characterise these enzymes. This will be crucial for understanding their mechanisms, predicting their efficacy, and engineering optimized enzymes for therapeutic applications.
Collapse
Affiliation(s)
- Max J Cheetham
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6AA, U.K
| | - Yunlong Huo
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6AA, U.K
| | - Maria Stroyakovski
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6AA, U.K
| | - Li Cheng
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6AA, U.K
| | - Daniel Wan
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6AA, U.K
| | - Anne Dell
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, U.K
| | - Joanne M Santini
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6AA, U.K
| |
Collapse
|
4
|
Khan S, Lin PR, Tan C. Engineering Cyborg Pathogens through Intracellular Hydrogelation. ACS Synth Biol 2024; 13:3609-3620. [PMID: 39413025 DOI: 10.1021/acssynbio.4c00420] [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: 10/18/2024]
Abstract
Synthetic biology primarily focuses on two kinds of cell chassis: living cells and nonliving systems. Living cells are autoreplicating systems that have active metabolism. Nonliving systems, including artificial cells and nanoparticles, are nonreplicating systems typically lacking active metabolism. In recent work, Cyborg bacteria that are nonreplicating-but-metabolically active have been engineered through intracellular hydrogelation. Intracellular hydrogelation is conducted by infusing gel monomers and photoactivators into cells, followed by the activation of polymerization of the gel monomers inside the cells. However, the previous work investigated only Escherichia coli cells. Extending the Cyborg-Cell method to pathogenic bacteria could enable the exploitation of their pathogenic properties in biomedical applications. Here, we focus on different strains of Pseudomonas aeruginosa, Staphylococcus aureus, and Klebsiella pneumoniae. To synthesize the Cyborg pathogens, we first reveal the impact of different hydrogel concentrations on the metabolism, replication, and intracellular gelation of Cyborg pathogens. Next, we demonstrate that the Cyborg pathogens are taken up by macrophages in a similar magnitude as wild-type pathogens through confocal microscopy and real-time PCR. Finally, we show that the macrophage that takes up the Cyborg pathogen exhibits a similar phenotypic response to the wild-type pathogen. Our work generalizes the intracellular hydrogelation approach from lab strains of E. coli to bacterial pathogens. The new Cyborg pathogens could be applied in biomedical applications ranging from drug delivery to immunotherapy.
Collapse
Affiliation(s)
- Shahid Khan
- Department of Biomedical Engineering, University of California, Davis, Davis, California 95616, United States
| | - Pin-Ru Lin
- Department of Biomedical Engineering, University of California, Davis, Davis, California 95616, United States
| | - Cheemeng Tan
- Department of Biomedical Engineering, University of California, Davis, Davis, California 95616, United States
| |
Collapse
|
5
|
Miller JC, Cross AS, Tennant SM, Baliban SM. Klebsiella pneumoniae Lipopolysaccharide as a Vaccine Target and the Role of Antibodies in Protection from Disease. Vaccines (Basel) 2024; 12:1177. [PMID: 39460343 PMCID: PMC11512408 DOI: 10.3390/vaccines12101177] [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: 08/16/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Klebsiella pneumoniae is well recognized as a serious cause of infection in healthcare-associated settings and immunocompromised individuals; however, accumulating evidence from resource-limited nations documents an alarming rise in community-acquired K. pneumoniae infections, manifesting as bacteremia and pneumonia as well as neonatal sepsis. The emergence of hypervirulent and antibiotic-resistant K. pneumoniae strains threatens treatment options for clinicians. Effective vaccination strategies could represent a viable alternative that would both preempt the need for antibiotics to treat K. pneumoniae infections and reduce the burden of K. pneumoniae disease globally. There are currently no approved K. pneumoniae vaccines. We review the evidence for K. pneumoniae lipopolysaccharide (LPS) as a vaccine and immunotherapeutic target and discuss the role of antibodies specific for the core or O-antigen determinants within LPS in protection against Klebsiella spp. disease. We expand on the known role of the Klebsiella spp. capsule and O-antigen modifications in antibody surface accessibility to LPS as well as the in vitro and in vivo effector functions reported for LPS-specific antibodies. We summarize key hypotheses stemming from these studies, review the role of humoral immunity against K. pneumoniae O-antigen for protection, and identify areas requiring further research.
Collapse
Affiliation(s)
- Jernelle C. Miller
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.C.M.); (A.S.C.); (S.M.T.)
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Alan S. Cross
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.C.M.); (A.S.C.); (S.M.T.)
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sharon M. Tennant
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.C.M.); (A.S.C.); (S.M.T.)
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Scott M. Baliban
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.C.M.); (A.S.C.); (S.M.T.)
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
6
|
Theodorakis N, Feretzakis G, Hitas C, Kreouzi M, Kalantzi S, Spyridaki A, Kollia Z, Verykios VS, Nikolaou M. Immunosenescence: How Aging Increases Susceptibility to Bacterial Infections and Virulence Factors. Microorganisms 2024; 12:2052. [PMID: 39458361 PMCID: PMC11510421 DOI: 10.3390/microorganisms12102052] [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: 09/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The process of aging leads to a progressive decline in the immune system function, known as immunosenescence, which compromises both innate and adaptive responses. This includes impairments in phagocytosis and decreased production, activation, and function of T- and B-lymphocytes, among other effects. Bacteria exploit immunosenescence by using various virulence factors to evade the host's defenses, leading to severe and often life-threatening infections. This manuscript explores the complex relationship between immunosenescence and bacterial virulence, focusing on the underlying mechanisms that increase vulnerability to bacterial infections in the elderly. Additionally, it discusses how machine learning methods can provide accurate modeling of interactions between the weakened immune system and bacterial virulence mechanisms, guiding the development of personalized interventions. The development of vaccines, novel antibiotics, and antivirulence therapies for multidrug-resistant bacteria, as well as the investigation of potential immune-boosting therapies, are promising strategies in this field. Future research should focus on how machine learning approaches can be integrated with immunological, microbiological, and clinical data to develop personalized interventions that improve outcomes for bacterial infections in the growing elderly population.
Collapse
Affiliation(s)
- Nikolaos Theodorakis
- Department of Cardiology, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
- School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527 Athens, Greece
| | - Georgios Feretzakis
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece;
| | - Christos Hitas
- Department of Cardiology, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
| | - Magdalini Kreouzi
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
- Department of Internal Medicine, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece
| | - Sofia Kalantzi
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
- Department of Internal Medicine, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece
| | - Aikaterini Spyridaki
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
- Department of Internal Medicine, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece
| | - Zoi Kollia
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
| | - Vassilios S. Verykios
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece;
| | - Maria Nikolaou
- Department of Cardiology, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
| |
Collapse
|
7
|
Chen Z, Gou Q, Yuan Y, Zhang X, Zhao Z, Liao J, Zeng X, Jing H, Jiang S, Zhang W, Zeng H, Huang W, Zou Q, Zhang J. Vaccination with a trivalent Klebsiella pneumoniae vaccine confers protection in a murine model of pneumonia. Vaccine 2024; 42:126217. [PMID: 39163713 DOI: 10.1016/j.vaccine.2024.126217] [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: 02/16/2024] [Revised: 07/01/2024] [Accepted: 08/08/2024] [Indexed: 08/22/2024]
Abstract
Klebsiella pneumoniae (K. pneumoniae) is an opportunistic pathogen and the major cause of healthcare-associated infections, which are increasingly complicated by the prevalence of highly invasive and hyper-virulent K. pneumoniae strains, necessitating the development of alternative strategies for combatting infections caused by this bacterium. In this study, we successfully constructed a fusion antigen called KP-Ag1, comprising three antigens (GlnH, FimA, and KPN_00466) that were previously identified through reverse vaccinology. Immunization with KP-Ag1 formulated with Al(OH)3 adjuvant elicited robust humoral and cellular immune response in mice, and conferred protective immunity in a murine model of K. pneumoniae lung infection. Further analysis of serum IgG subtypes from mice immunized with KP-Ag1 revealed a predominant IgG1 response, indicating that KP-Ag1 predominantly induces a Th2-biased immune response. Additionally, opsonophagocytic killing assay suggested that humoral immune responses play a pivotal role in mediating protection conferred by KP-Ag1. Moreover, KP-Ag1 was found to promote the activation and maturation of BMDCs in vitro, which is essential for subsequent efficient antigen presentation. More importantly, vaccination with KP-Ag1 demonstrated cross-protective efficacy against clinical isolates of K. pneumoniae varying in serotypes, antibiotic resistance, and virulence profiles. Therefore, KP-Ag1 holds promise as a candidate for K. pneumoniae vaccine development.
Collapse
MESH Headings
- Animals
- Klebsiella pneumoniae/immunology
- Klebsiella Infections/prevention & control
- Klebsiella Infections/immunology
- Mice
- Disease Models, Animal
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/immunology
- Bacterial Vaccines/immunology
- Bacterial Vaccines/administration & dosage
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Adjuvants, Immunologic/administration & dosage
- Female
- Immunity, Humoral
- Vaccination/methods
- Antigens, Bacterial/immunology
- Pneumonia, Bacterial/prevention & control
- Pneumonia, Bacterial/immunology
- Mice, Inbred BALB C
- Immunity, Cellular
- Cross Protection/immunology
Collapse
Affiliation(s)
- Zhifu Chen
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Qiang Gou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Yue Yuan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Xiaoli Zhang
- Department of Clinical Hematology, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Zhuo Zhao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Jingwen Liao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Xi Zeng
- Department of Pharmacy, General Hospital of Northern Theatre Command, Shenyang 110016, PR China
| | - Haiming Jing
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Shichun Jiang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Weijun Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Wei Huang
- Department of Medical Laboratory, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, PR China.
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China.
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China.
| |
Collapse
|
8
|
Dey J, Mahapatra SR, Raj TK, Misra N, Suar M. Identification of potential flavonoid compounds as antibacterial therapeutics against Klebsiella pneumoniae infection using structure-based virtual screening and molecular dynamics simulation. Mol Divers 2024; 28:3111-3128. [PMID: 37801217 DOI: 10.1007/s11030-023-10738-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/25/2023] [Indexed: 10/07/2023]
Abstract
Klebsiella pneumoniae, which is among the top three pathogens on WHO's priority list, is one of the gram-negative bacteria that doctors and researchers around the world have fought for decades. Capsular polysaccharide (CPS) protein is extensively recognized as an important K. pneumoniae virulence factor. Thus, CPS has become the most characterized target for the discovery of novel drug candidates. The ineffectiveness of currently existing antibiotics urges the search for potent antimicrobial compounds. Flavonoids are a group of plant metabolites that have antibacterial potential and can enhance the present medications to elicit improved results against diverse diseases without adverse reactions. Henceforth, the present study aims to illustrate the inhibitory potential of flavonoids with varying pharmacological properties, targeting the CPS protein of K. pneumoniae by in silico approaches. The flavonoid compounds (n = 169) were retrieved from the PubChem database and screened using the structure-based virtual screening approach. Compounds with the highest binding score were estimated through their pharmacokinetic effects by ADMET descriptors. Finally, four potential inhibitors with PubChem CID: (4301534, 5213, 5481948, and 637080) were selected after molecular docking and drug-likeness analysis. All four lead compounds were employed for the MDS analysis of a 100 ns time period. Various studies were undertaken to assess the stability of the protein-ligand complexes. The binding free energy was computed using MM-PBSA, and the outcomes indicated that the molecules are having stable interactions with the binding site of the target protein. The results revealed that all four compounds can be employed as potential therapeutics against K. pneumoniae.
Collapse
Affiliation(s)
- Jyotirmayee Dey
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, India
| | - Soumya Ranjan Mahapatra
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, India
| | - T Kiran Raj
- Department of Biotechnology & Bioinformatics, School of Life Sciences, JSS Academy of Higher Education & Research, Mysore, India
| | - Namrata Misra
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, India.
- KIIT-Technology Business Incubator (KIIT-TBI), Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, India.
| | - Mrutyunjay Suar
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, India.
- KIIT-Technology Business Incubator (KIIT-TBI), Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, India.
| |
Collapse
|
9
|
Schmidt S, Mondino S, Gomez-Valero L, Escoll P, Mascarenhas DPA, Gonçalves A, Camara PHM, Garcia Rodriguez FJ, Rusniok C, Sachse M, Moya-Nilges M, Fontaine T, Zamboni DS, Buchrieser C. The unique Legionella longbeachae capsule favors intracellular replication and immune evasion. PLoS Pathog 2024; 20:e1012534. [PMID: 39259722 PMCID: PMC11419355 DOI: 10.1371/journal.ppat.1012534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 09/23/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024] Open
Abstract
Legionella longbeachae and Legionella pneumophila are the most common causative agents of Legionnaires' disease. While the clinical manifestations caused by both species are similar, species-specific differences exist in environmental niches, disease epidemiology, and genomic content. One such difference is the presence of a genomic locus predicted to encode a capsule. Here, we show that L. longbeachae indeed expresses a capsule in post-exponential growth phase as evidenced by electron microscopy analyses, and that capsule expression is abrogated when deleting a capsule transporter gene. Capsule purification and its analysis via HLPC revealed the presence of a highly anionic polysaccharide that is absent in the capsule mutant. The capsule is important for replication and virulence in vivo in a mouse model of infection and in the natural host Acanthamoeba castellanii. It has anti-phagocytic function when encountering innate immune cells such as human macrophages and it is involved in the low cytokine responses in mice and in human monocyte derived macrophages, thus dampening the innate immune response. Thus, the here characterized L. longbeachae capsule is a novel virulence factor, unique among the known Legionella species, which may aid L. longbeachae to survive in its specific niches and which partly confers L. longbeachae its unique infection characteristics.
Collapse
Affiliation(s)
- Silke Schmidt
- Institut Pasteur, Université Paris Cité, Biologie des Bactéries Intracellulaires, CNRS UMR 6047, Paris, France
- Sorbonne Université, Collège Doctoral, Paris, France
| | - Sonia Mondino
- Institut Pasteur, Université Paris Cité, Biologie des Bactéries Intracellulaires, CNRS UMR 6047, Paris, France
| | - Laura Gomez-Valero
- Institut Pasteur, Université Paris Cité, Biologie des Bactéries Intracellulaires, CNRS UMR 6047, Paris, France
| | - Pedro Escoll
- Institut Pasteur, Université Paris Cité, Biologie des Bactéries Intracellulaires, CNRS UMR 6047, Paris, France
| | | | - Augusto Gonçalves
- Department of Cell Biology, Medical School of Ribeirão Preto, FMRP/USP, Ribeirão Preto, Brazil
| | - Pedro H. M. Camara
- Department of Cell Biology, Medical School of Ribeirão Preto, FMRP/USP, Ribeirão Preto, Brazil
| | | | - Christophe Rusniok
- Institut Pasteur, Université Paris Cité, Biologie des Bactéries Intracellulaires, CNRS UMR 6047, Paris, France
| | - Martin Sachse
- UTechS UBI, Centre de Ressources et Recherches Technologiques, Institut Pasteur, Paris, France
| | - Maryse Moya-Nilges
- UTechS UBI, Centre de Ressources et Recherches Technologiques, Institut Pasteur, Paris, France
| | - Thierry Fontaine
- Biologie et Pathogénicité fongiques, Institut Pasteur, Paris, France
| | - Dario S. Zamboni
- Department of Cell Biology, Medical School of Ribeirão Preto, FMRP/USP, Ribeirão Preto, Brazil
| | - Carmen Buchrieser
- Institut Pasteur, Université Paris Cité, Biologie des Bactéries Intracellulaires, CNRS UMR 6047, Paris, France
| |
Collapse
|
10
|
El-Demerdash AS, Alfaraj R, Farid FA, Yassin MH, Saleh AM, Dawwam GE. Essential oils as capsule disruptors: enhancing antibiotic efficacy against multidrug-resistant Klebsiella pneumoniae. Front Microbiol 2024; 15:1467460. [PMID: 39282565 PMCID: PMC11392748 DOI: 10.3389/fmicb.2024.1467460] [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: 07/19/2024] [Accepted: 08/13/2024] [Indexed: 09/19/2024] Open
Abstract
Background Multidrug-resistant Klebsiella pneumoniae (MDR-KP) poses a significant global health threat due to its involvement in severe infections and high mortality rates. The emergence of MDR strains necessitates the exploration of alternative therapeutic strategies. Methods K. pneumoniae isolates were obtained from human and animal sources. Antibacterial susceptibility testing was performed, followed by the evaluation of essential oil activity through inhibition zone, MIC, and MBC determinations. Checkerboard assays were conducted to assess synergistic effects with amikacin. Gene expression analysis and transmission electron microscopy were employed to elucidate the mechanisms of action. Molecular docking studies were performed to identify potential binding targets of bioactive compounds. Results Klebsiella pneumoniae was isolated from 25 of the100 samples examined, representing a prevalence rate of 25%. All isolates were found to be multidrug-resistant. Tea tree and thyme essential oils exhibited potent antibacterial activity and synergistic effects with amikacin. Notably, these combinations significantly downregulated the expression of key capsule virulence genes (wcaG, rmpA, magA, uge, and wabG), suggesting a novel mechanism for enhancing amikacin efficacy. Transmission electron microscopy revealed disrupted cell integrity in MDR-KP cells treated with the combinations. Molecular docking analysis identified Terpinen-4-ol, Farnesol, 1,4-Dihydroxy-p-menth-2-ene, and 7-Oxabicyclo [4.1.0] heptane as potential bioactive compounds responsible for the observed effects. Conclusion By effectively combating MDR-KP, this research holds promise for reducing antibiotic resistance, improving treatment outcomes, and ultimately enhancing potential care.
Collapse
Affiliation(s)
- Azza SalahEldin El-Demerdash
- Laboratory of Biotechnology, Department of Microbiology, Agricultural Research Center (ARC), Animal Health Research Institute (AHRI), Zagazig, Egypt
| | - Rihaf Alfaraj
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Faten A Farid
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha, Egypt
| | - Mohamed H Yassin
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha, Egypt
| | - Abdulrahman M Saleh
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
- Epidemiological Surveillance Unit, Aweash El-Hagar Family Medicine Center, MOHP, Mansoura, Egypt
| | - Ghada E Dawwam
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha, Egypt
| |
Collapse
|
11
|
Bain W, Ahn B, Peñaloza HF, McElheny CL, Tolman N, van der Geest R, Gonzalez-Ferrer S, Chen N, An X, Hosuru R, Tabary M, Papke E, Kohli N, Farooq N, Bachman W, Olonisakin TF, Xiong Z, Griffith MP, Sullivan M, Franks J, Mustapha MM, Iovleva A, Suber T, Shanks RQ, Ferreira VP, Stolz DB, Van Tyne D, Doi Y, Lee JS. In Vivo Evolution of a Klebsiella pneumoniae Capsule Defect With wcaJ Mutation Promotes Complement-Mediated Opsonophagocytosis During Recurrent Infection. J Infect Dis 2024; 230:209-220. [PMID: 39052750 PMCID: PMC11272070 DOI: 10.1093/infdis/jiae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/17/2023] [Accepted: 01/03/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Klebsiella pneumoniae carbapenemase-producing K pneumoniae (KPC-Kp) bloodstream infections are associated with high mortality. We studied clinical bloodstream KPC-Kp isolates to investigate mechanisms of resistance to complement, a key host defense against bloodstream infection. METHODS We tested growth of KPC-Kp isolates in human serum. In serial isolates from a single patient, we performed whole genome sequencing and tested for complement resistance and binding by mixing study, direct enzyme-linked immunosorbent assay, flow cytometry, and electron microscopy. We utilized an isogenic deletion mutant in phagocytosis assays and an acute lung infection model. RESULTS We found serum resistance in 16 of 59 (27%) KPC-Kp clinical bloodstream isolates. In 5 genetically related bloodstream isolates from a single patient, we noted a loss-of-function mutation in the capsule biosynthesis gene, wcaJ. Disruption of wcaJ was associated with decreased polysaccharide capsule, resistance to complement-mediated killing, and surprisingly, increased binding of complement proteins. Furthermore, an isogenic wcaJ deletion mutant exhibited increased opsonophagocytosis in vitro and impaired in vivo control in the lung after airspace macrophage depletion in mice. CONCLUSIONS Loss of function in wcaJ led to increased complement resistance, complement binding, and opsonophagocytosis, which may promote KPC-Kp persistence by enabling coexistence of increased bloodstream fitness and reduced tissue virulence.
Collapse
Affiliation(s)
- William Bain
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Brian Ahn
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus School of Medicine, Denver
| | - Hernán F Peñaloza
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | | | - Nathanial Tolman
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Rick van der Geest
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Shekina Gonzalez-Ferrer
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Nathalie Chen
- Division of Infectious Diseases, Department of Medicine
| | - Xiaojing An
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Ria Hosuru
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Mohammadreza Tabary
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Erin Papke
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Naina Kohli
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | | | | | - Tolani F Olonisakin
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Zeyu Xiong
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | | | - Mara Sullivan
- Center for Biologic Imaging, Department of Cell Biology
| | | | | | - Alina Iovleva
- Division of Infectious Diseases, Department of Medicine
| | - Tomeka Suber
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
| | - Robert Q Shanks
- Department of Ophthalmology, University of Pittsburgh, Pennsylvania
| | - Viviana P Ferreira
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Ohio
| | - Donna B Stolz
- Center for Biologic Imaging, Department of Cell Biology
| | | | - Yohei Doi
- Division of Infectious Diseases, Department of Medicine
| | - Janet S Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh
- Division of Pulmonary and Critical Care Medicine, Washington University in St Louis, Missouri
| |
Collapse
|
12
|
Shamanna V, Srinivas S, Couto N, Nagaraj G, Sajankila SP, Krishnappa HG, Kumar KA, Aanensen DM, Lingegowda RK. Geographical distribution, disease association and diversity of Klebsiella pneumoniae K/L and O antigens in India: roadmap for vaccine development. Microb Genom 2024; 10:001271. [PMID: 39037209 PMCID: PMC11316559 DOI: 10.1099/mgen.0.001271] [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: 03/26/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024] Open
Abstract
Klebsiella pneumoniae poses a significant healthcare challenge due to its multidrug resistance and diverse serotype landscape. This study aimed to explore the serotype diversity of 1072 K. pneumoniae and its association with geographical distribution, disease severity and antimicrobial/virulence patterns in India. Whole-genome sequencing was performed on the Illumina platform, and genomic analysis was carried out using the Kleborate tool. The analysis revealed a total of 78 different KL types, among which KL64 (n=274/1072, 26 %), KL51 (n=249/1072, 24 %), and KL2 (n=88/1072, 8 %) were the most prevalent. In contrast, only 13 distinct O types were identified, with O1/O2v1 (n=471/1072, 44 %), O1/O2v2 (n=353/1072, 33 %), and OL101 (n=66/1072, 6 %) being the predominant serotypes. The study identified 114 different sequence types (STs) with varying serotypes, with ST231 being the most predominant. O serotypes were strongly linked with STs, with O1/O2v1 predominantly associated with ST231. Simpson's diversity index and Fisher's exact test revealed higher serotype diversity in the north and east regions, along with intriguing associations between specific serotypes and resistance profiles. No significant association between KL or O types and disease severity was observed. Furthermore, we found the specific association of virulence factors yersiniabactin and aerobactin (P<0.05) with KL types but no association with O antigen types (P>0.05). Conventionally described hypervirulent clones (i.e. KL1 and KL2) in India lacked typical virulent markers (i.e. aerobactin), contrasting with other regional serotypes (KL51). The cumulative distribution of KL and O serotypes suggests that future vaccines may have to include either ~20 KL or four O types to cover >85 % of the carbapenemase-producing Indian K. pneumoniae population. The results highlight the necessity for comprehensive strategies to manage the diverse landscape of K. pneumoniae strains across different regions in India. Understanding regional serotype dynamics is pivotal for targeted surveillance, interventions, and tailored vaccine strategies to tackle the diverse landscape of K. pneumoniae infections across India. This article contains data hosted by Microreact.
Collapse
Affiliation(s)
- Varun Shamanna
- Central Research Laboratory, KIMS, Bengaluru, India
- Department of Biotechnology, NMAM Institute of Technology, Nitte, Udupi, India
| | | | - Natacha Couto
- Centre for Genomic Pathogen Surveillance, Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | | | | | | | | | - David M. Aanensen
- Centre for Genomic Pathogen Surveillance, Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | | | - NIHR Global Health Research Unit on genomic surveillance - India consortium
- Central Research Laboratory, KIMS, Bengaluru, India
- Department of Biotechnology, NMAM Institute of Technology, Nitte, Udupi, India
- Centre for Genomic Pathogen Surveillance, Pandemic Sciences Institute, University of Oxford, Oxford, UK
| |
Collapse
|
13
|
Abu-Aqil G, Suleiman M, Lapidot I, Huleihel M, Salman A. Infrared spectroscopy-based machine learning algorithms for rapid detection of Klebsiella pneumoniae isolated directly from patients' urine and determining its susceptibility to antibiotics. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 314:124141. [PMID: 38513317 DOI: 10.1016/j.saa.2024.124141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/15/2024] [Accepted: 03/08/2024] [Indexed: 03/23/2024]
Abstract
Among the most prevalent and detrimental bacteria causing urinary tract infections (UTIs) is Klebsiella (K.) pneumoniae. A rapid determination of its antibiotic susceptibility can enhance patient treatment and mitigate the spread of resistant strains. In this study, we assessed the viability of using infrared spectroscopy-based machine learning as a rapid and precise approach for detecting K. pneumoniae bacteria and determining its susceptibility to various antibiotics directly from a patient's urine sample. In this study, 2333 bacterial samples, including 636 K. pneumoniae were investigated using infrared micro-spectroscopy. The obtained spectra (27996spectra) were analyzed with XGBoost classifier, achieving a success rate exceeding 95 % for identifying K. pneumoniae. Moreover, this method allows for the simultaneous determination of K. pneumoniae susceptibility to various antibiotics with sensitivities ranging between 74 % and 81 % within approximately 40 min after receiving the patient's urine sample.
Collapse
Affiliation(s)
- George Abu-Aqil
- Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Manal Suleiman
- Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Itshak Lapidot
- Department of Electrical and Electronics Engineering, ACLP-Afeka Center for Language Processing, Afeka Tel-Aviv Academic College of Engineering, Tel-Aviv 69107, Israel
| | - Mahmoud Huleihel
- Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| | - Ahmad Salman
- Department of Physics, SCE - Shamoon College of Engineering, Beer-Sheva 84100, Israel.
| |
Collapse
|
14
|
de Melo AG, Morency C, Moineau S. Virulence-associated factors as targets for phage infection. Curr Opin Microbiol 2024; 79:102471. [PMID: 38569419 DOI: 10.1016/j.mib.2024.102471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/05/2024]
Abstract
Bacterial pathogens can infect a wide range of hosts and pose a threat to public and animal health as well as to agriculture. The emergence of antibiotic-resistant strains has increased this risk by making the treatment of bacterial infections even more challenging. Pathogenic bacteria thrive in various ecological niches, but they can also be specifically targeted and killed by bacteriophages (phages). Lytic phages are now investigated and even used, in some cases, as alternatives or complements to antibiotics for preventing or treating bacterial infections (phage therapy). As such, it is key to identify factors responsible for phage specificity and efficiency. Here, we review recent advances in virulence-associated factors that are targeted by phages. We highlight components of the bacterial cell surface, effector systems, and motility structures exploited by phages and the effects of phages on cell aggregation and communication. We also look at the fitness trade-off of phage resistance.
Collapse
Affiliation(s)
- Alessandra G de Melo
- Département de biochimie, de microbiologie, et de bio-informatique, Faculté des sciences et de génie, Québec City, QC G1V 0A6, Canada
| | - Carlee Morency
- Département de biochimie, de microbiologie, et de bio-informatique, Faculté des sciences et de génie, Québec City, QC G1V 0A6, Canada
| | - Sylvain Moineau
- Département de biochimie, de microbiologie, et de bio-informatique, Faculté des sciences et de génie, Québec City, QC G1V 0A6, Canada; Félix d'Hérelle Reference Center for Bacterial Viruses, Université Laval, Québec City, QC G1V 0A6, Canada.
| |
Collapse
|
15
|
Huang T, Zhang Z, Tao X, Shi X, Lin P, Liao D, Ma C, Cai X, Lin W, Jiang X, Luo P, Wu S, Xie Y. Structural and functional basis of bacteriophage K64-ORF41 depolymerase for capsular polysaccharide degradation of Klebsiella pneumoniae K64. Int J Biol Macromol 2024; 265:130917. [PMID: 38513899 DOI: 10.1016/j.ijbiomac.2024.130917] [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: 01/17/2024] [Revised: 03/04/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024]
Abstract
Capsule polysaccharide is an important virulence factor of Klebsiella pneumoniae (K. pneumoniae), which protects bacteria against the host immune response. A promising therapeutic approach is using phage-derived depolymerases to degrade the capsular polysaccharide and expose and sensitize the bacteria to the host immune system. Here we determined the cryo-electron microscopy (cryo-EM) structures of a bacteriophage tail-spike protein against K. pneumoniae K64, ORF41 (K64-ORF41) and ORF41 in EDTA condition (K64-ORF41EDTA), at 2.37 Å and 2.50 Å resolution, respectively, for the first time. K64-ORF41 exists as a trimer and each protomer contains a β-helix domain including a right-handed parallel β-sheet helix fold capped at both ends, an insertion domain, and one β-sheet jellyroll domain. Moreover, our structural comparison with other depolymerases of K. pneumoniae suggests that the catalytic residues (Tyr528, His574 and Arg628) are highly conserved although the substrate of capsule polysaccharide is variable. Besides that, we figured out the important residues involved in the substrate binding pocket including Arg405, Tyr526, Trp550 and Phe669. This study establishes the structural and functional basis for the promising phage-derived broad-spectrum activity depolymerase therapeutics and effective CPS-degrading agents for the treatment of carbapenem-resistant K. pneumoniae K64 infections.
Collapse
Affiliation(s)
- Tianyun Huang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Department of Pharmacy, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710021, China; School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zhuoyuan Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Xin Tao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Xinyu Shi
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Peng Lin
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Dan Liao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Chenyu Ma
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xinle Cai
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Wei Lin
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Shan Wu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China.
| | - Yuan Xie
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
16
|
Qazi F, Verma R, Redmond CE, Khalid A, O'Brien-Simpson NM, Tomljenovic-Hanic S. Real-time, label-free detection and identification of bacteria through non-invasive optical imaging. Microbes Infect 2024; 26:105263. [PMID: 38013067 DOI: 10.1016/j.micinf.2023.105263] [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: 04/27/2023] [Revised: 11/14/2023] [Accepted: 11/18/2023] [Indexed: 11/29/2023]
Abstract
Currently, traditional and newer molecular and mass spectrometry techniques of identifying bacteria from biological samples requires lengthy sample preparation, growth and labelling/staining assays. Thus, there is a pressing clinical need for an adjunct method that accurately identifies bacteria in real time. Here we report on the evaluation of confocal microscopy for the identification of clinically important and multi-drug resistant (MDR) bacteria in real time, using their intrinsic fluorescence features, i.e., emission spectra and fluorescence lifetime. The results demonstrate that difference in emission spectra and fluorescence lifetimes can be used as a fingerprint for identification of 12 bacterial species and MDR strains in real-time. Photostability or time-traces of bacteria demonstrated that these parameters could be used for tracking and recording without a need for labelling. Further, dilution experiments demonstrated that using intrinsic fluorescence S. aureus, Klebsiella pneumoniae and Escherichia coli bacteria can be detected and identified at clinically relevant concentrations as low as 2 × 102 CFU/mL. This non-invasive, non-labelling optical methodology may serve as the basis for development of a device that would quickly and accurately identify bacteria in biological samples. Thus, this intrinsic fluorescence technique would provide clinicians information, within minutes from sampling, to base accurate and specific treatments for patients.
Collapse
Affiliation(s)
- Farah Qazi
- School of Physics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Rajni Verma
- School of Physics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Connagh E Redmond
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, University of Melbourne, Melbourne, Victoria, 3010, Australia
| | - Asma Khalid
- School of Science, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Neil M O'Brien-Simpson
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, University of Melbourne, Melbourne, Victoria, 3010, Australia
| | | |
Collapse
|
17
|
Lahij HF, Almeani SAL. Multidrug-resistant clinical K. pneumoniae ST16, ST218, and ST283 and emergence of pandrug-resistant KPC-positive ST6434/K2 lineage in Iraq. Braz J Microbiol 2024; 55:375-382. [PMID: 38091237 PMCID: PMC10920612 DOI: 10.1007/s42770-023-01205-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/28/2023] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND AND AIM The increasing incidence of Klebsiella pneumoniae infections, both in the community and in hospitals, is a huge health problem. This is due to the increasing resistance of the bacteria to antibiotics and biofilm formation, as well as the presence of a capsule. This study focuses on two main objectives: to survey the most common capsular types in local isolates for the first time in Anbar, Iraq, on molecular level and to distinguish between infectious pathogen strains using multilocus sequence typing (MLST) for more efficient epidemiological and surveillance analysis, in order to determine the source of these strains (invasive or purebred). METHODOLOGY Multidrug-resistant (MDR) isolates adapted to genomic extraction and molecular screening of capsular type and MLST, and then to data processing by Pasteur Institut. RESULTS For the first time, one isolate was registered as a new strain in the world with ST 6434; the other strains demonstrated as preregistered with ST16, ST218, and ST283. 33% of MDR isolates belonged to the capsular K2 type. CONCLUSION The study's findings were not aligned with the global knowledge base about the distribution of capsular type in Asia. To prevent the spread of highly resistant strains, careful monitoring of virulence determinants is necessary in addition to the observation of antibiotic resistance.
Collapse
Affiliation(s)
- Hasan Falah Lahij
- Department of Biology, Collage of Sciences, University of Anbar, Ramadi, Anbar, Iraq.
- Medical Laboratory Technology, Almaarif University College, Ramadi, 31001, Iraq.
| | | |
Collapse
|
18
|
Liu N, Lou N, Huang J, Chen Z, Li B, Zhang Z, Hong Y, Cao L, Xiao W. Genomic surveillance indicates clonal replacement of hypervirulent Klebsiella pneumoniae ST881 and ST29 lineage strains in vivo. Front Microbiol 2024; 15:1375624. [PMID: 38440138 PMCID: PMC10910047 DOI: 10.3389/fmicb.2024.1375624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 02/08/2024] [Indexed: 03/06/2024] Open
Abstract
The emergence of hypervirulent Klebsiella pneumoniae (hvKp) poses a significant public health threat, particularly regarding its carriage in the healthy population. However, the genomic epidemiological characteristics and population dynamics of hvKp within a single patient across distinct infection episodes remain largely unknown. This study aimed to investigate the clonal replacement of hvKp K2-ST881 and K54-ST29 lineage strains in a single patient experiencing multiple-site infections during two independent episodes. Two strains, designated EDhvKp-1 and EDhvKp-2, were obtained from blood and cerebrospinal fluid during the first admission, and the strain isolated from blood on the second admission was named EDhvKp-3. Whole-genome sequencing, utilizing both short-read Illumina and long-read Oxford Nanopore platforms, was conducted. In silico multilocus sequence typing (MLST), identification of antimicrobial resistance and virulence genes, and the phylogenetic relationship between our strains and other K. pneumoniae ST881 and ST29 genomes retrieved from the public database were performed. Virulence potentials were assessed through a mouse lethality assay. Our study indicated that the strains were highly susceptible to multiple antimicrobial agents. Plasmid sequence analysis confirmed that both virulence plasmids, pEDhvKp-1 (166,008 bp) and pEDhvKp-3 (210,948 bp), belonged to IncFIB type. Multiple virulence genes, including rmpA, rmpA2, rmpC, rmpD, iroBCDN, iucABCD, and iutA, were identified. EDhvKp-1 and EDhvKp-2 showed the closest relationship to strain 502 (differing by 51 SNPs), while EDhvKp-3 exhibited 69 SNPs differences compared to strain TAKPN-1, which all recovered from Chinese patients in 2020. In the mouse infection experiment, both ST881 EDhvKp-1 and ST29 EDhvKp-3 displayed similar virulence traits, causing 90 and 100% of the mice to die within 72 h after intraperitoneal infection, respectively. Our study expands the spectrum of hvKp lineages and highlights genomic alterations associated with clonal switching between two distinct lineages of hvKP that successively replaced each other in vivo. The development of novel strategies for the surveillance, diagnosis, and treatment of high-risk hvKp is urgently needed.
Collapse
Affiliation(s)
- Ning Liu
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ningjie Lou
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiajie Huang
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenhao Chen
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing Li
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhongheng Zhang
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yucai Hong
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liping Cao
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Xiao
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
19
|
Luna-Pineda VM, Rodríguez-Martínez G, Salazar-García M, Romo-Castillo M. Plant-Origin Components: New Players to Combat Antibiotic Resistance in Klebsiella pneumoniae. Int J Mol Sci 2024; 25:2134. [PMID: 38396811 PMCID: PMC10888558 DOI: 10.3390/ijms25042134] [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: 12/01/2023] [Revised: 01/09/2024] [Accepted: 01/13/2024] [Indexed: 02/25/2024] Open
Abstract
Klebsiella pneumoniae (Kpn) is an opportunistic pathogen that causes intrahospital complications such as pneumonia, liver abscesses, soft tissue infections, urinary infections, bacteraemia, and, in some cases, death. Since this bacterium has a higher frequency than other Gram-negative pathogens, it has become an important pathogen to the health sector. The adaptative genome of Kpn likely facilitates increased survival of the pathogen in diverse situations. Therefore, several studies have been focused on developing new molecules, synergistic formulations, and biomaterials that make it possible to combat and control infections with and dispersion of this pathogen. Note that the uncontrolled antibiotic administration that occurred during the pandemic led to the emergence of new multidrug-resistant strains, and scientists were challenged to overcome them. This review aims to compile the latest information on Kpn that generates intrahospital infections, specifically their pathogenicity-associated factors. Furthermore, it explains the natural-product-based treatments (extracts and essential oils) developed for Kpn infection and dispersion control.
Collapse
Affiliation(s)
- Victor M. Luna-Pineda
- Laboratorio de Investigación en COVID-19, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico; (V.M.L.-P.); (G.R.-M.)
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico
| | - Griselda Rodríguez-Martínez
- Laboratorio de Investigación en COVID-19, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico; (V.M.L.-P.); (G.R.-M.)
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico
| | - Marcela Salazar-García
- Departamento de Investigación Biomédica, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico;
| | - Mariana Romo-Castillo
- IxM/CONAHCYT-HIMFG, Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, Mexico
| |
Collapse
|
20
|
Bakhtiyari N, Farajnia S, Ghasemali S, Farajnia S, Pormohammad A, Saeidvafa S. Strategies to Overcome Antimicrobial Resistance in Nosocomial Infections, A Review and Update. Infect Disord Drug Targets 2024; 24:e260124226226. [PMID: 38284691 DOI: 10.2174/0118715265276529231214105423] [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: 08/26/2023] [Revised: 11/12/2023] [Accepted: 11/17/2023] [Indexed: 01/30/2024]
Abstract
Nosocomial infections, also known as healthcare-associated infections, are a significant global concern due to their strong association with high mortality and morbidity in both developed and developing countries. These infections are caused by a variety of pathogens, particularly the ESKAPE group of bacteria, which includes the six pathogens Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp. These bacteria have demonstrated noteworthy resistance to different antibiotics. Antimicrobial resistance mechanisms can manifest in various forms, including restricting drug uptake, modifying drug targets, inactivating drugs, active drug efflux, and biofilm formation. Accordingly, various strategies have been developed to combat antibiotic-resistant bacteria. These strategies encompass the development of new antibiotics, the utilization of bacteriophages that specifically target these bacteria, antimicrobial combination therapy and the use of peptides or enzymes that target the genomes or essential proteins of resistant bacteria. Among promising approaches to overcome antibiotic resistance, the CRISPR/Cas system stands out and offers many advantages. This system enables precise and efficient editing of genetic material at specific locations in the genome. Functioning as a bacterial "adaptive immune system," the CRISPR/Cas system recognizes, degrades, and remembers foreign DNA sequences through the use of spacer DNA segments that are transcribed into CRISPR RNAs (crRNA). This paper has focused on nosocomial infections, specifically the pathogens involved in hospital infections, the mechanisms underlying bacterial resistance, and the strategies currently employed to address this issue. Special emphasis has been placed on the application of CRISPR/Cas technology for overcoming antimicrobial resistance.
Collapse
Affiliation(s)
- Nasim Bakhtiyari
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safar Farajnia
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samaneh Ghasemali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Farajnia
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Pormohammad
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | | |
Collapse
|
21
|
Concha-Eloko R, Barberán-Martínez P, Sanjuán R, Domingo-Calap P. Broad-range capsule-dependent lytic Sugarlandvirus against Klebsiella sp. Microbiol Spectr 2023; 11:e0429822. [PMID: 37882584 PMCID: PMC10714931 DOI: 10.1128/spectrum.04298-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 09/15/2023] [Indexed: 10/27/2023] Open
Abstract
IMPORTANCE The emergence of multi-drug resistant bacteria is a global health problem. Among them, Klebsiella pneumoniae is considered a high-priority pathogen, making it necessary to develop new therapeutic tools to reduce the bacterial burden in an effective and sustainable manner. Phages, bacterial viruses, are very promising tools. However, phages are highy specific, rendering large-scale therapeutics costly to implement. This is especially certain in Klebsiella, a capsular bacterium in which phages have been shown to be capsular type dependent, infecting one or a few capsular types through specific enzymes called depolymerases. In this study, we have isolated and characterized novel phages with lytic ability against bacteria from a wide variety of capsular types, representing the Klebsiella phages with the widest range of infection described. Remarkably, these broad-range phages showed capsule dependency, despite the absence of depolymerases in their genomes, implying that infectivity could be governed by alternative mechanisms yet to be uncovered.
Collapse
Affiliation(s)
- Robby Concha-Eloko
- Instituto de Biología Integrativa de Sistemas, Universitat de València-CSIC, Paterna, Spain
| | | | - Rafael Sanjuán
- Instituto de Biología Integrativa de Sistemas, Universitat de València-CSIC, Paterna, Spain
| | - Pilar Domingo-Calap
- Instituto de Biología Integrativa de Sistemas, Universitat de València-CSIC, Paterna, Spain
| |
Collapse
|
22
|
da Cunha KF, de Oliveira Garcia M, Allend SO, de Albernaz DTF, da Rosa BN, Pereira IL, de Pereira de Pereira CM, Hartwig DD. Antibacterial and antibiofilm activity of 1-thiocarbamoyl-3,5-diaryl-4,5-dihydro-1H pyrazoles and thiazoles in multidrug-resistant pathogens. Braz J Microbiol 2023; 54:2587-2595. [PMID: 37656404 PMCID: PMC10689707 DOI: 10.1007/s42770-023-01110-2] [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: 03/17/2023] [Accepted: 08/07/2023] [Indexed: 09/02/2023] Open
Abstract
To find novel antibiotic drugs, six 1-thiocarbamoyl-3,5-diaryl-4,5-dihydro-1H derivatives named 1b, 1d (pyrazoles), 2a, 2b, 2c, and 2d (thiazoles) were evaluated in silico and in vitro. The in silico analyses were based on ADME pharmacokinetic parameters (absorption, distribution, metabolism, and excretion). The in vitro antibacterial activity was evaluated in Gram-positive and Gram-negative species (Staphylococcus aureus ATCC® 25904, Staphylococcus epidermidis ATCC® 35984, Klebsiella pneumoniae ATCC® 700603, and Acinetobacter baumannii ATCC® 19606), by determination of minimal inhibitory concentration (MIC), minimal bactericidal concentration (MBC), kinetics curve, and antibiofilm assays. As results, the azoles have activity against the Gram-negative species K. pneumoniae ATCC® 700603 and A. baumannii ATCC® 19606. No antibacterial activity was observed for the Gram-positive bacteria evaluated. Thus, the azoles were evaluated against clinical isolates of K. pneumoniae carbapenemase (KPC) and A. baumannii multidrug-resistant (Ab-MDR). All azoles have antibacterial activity against Ab-MDR isolates (Gram-negative) with MIC values between 512 μg/mL and 1,024 μg/mL. Against KPC isolates the azoles 1b, 1d, and 2d present antibacterial activity (MIC = 1,024 μg/mL). In the kinetics curve assay, the 1b and 1d pyrazoles reduced significantly viable cells of Ab-MDR isolates and additionally inhibited 86.6 to 95.8% of the biofilm formation. The in silico results indicate high possibility to permeate the blood-brain barrier (2b) and was predict human gastrointestinal absorption (all evaluated azoles). Considering that the research and development of new antibiotics is a priority for drug-resistant pathogens, our study revealed the antibacterial and antibiofilm activity of novel azoles against K. pneumoniae and A. baumannii pathogens.
Collapse
Affiliation(s)
- Kamila Furtado da Cunha
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, CEP: 96010-900, Brazil
| | - Marcelle de Oliveira Garcia
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, CEP: 96010-900, Brazil
| | - Suzane Olachea Allend
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, CEP: 96010-900, Brazil
- Biotechnology Unit, Technology Development Center, Federal University of Pelotas, Pelotas, RS, CEP: 96010-900, Brazil
| | - Déborah Trota Farias de Albernaz
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, CEP: 96010-900, Brazil
- Biotechnology Unit, Technology Development Center, Federal University of Pelotas, Pelotas, RS, CEP: 96010-900, Brazil
| | - Bruno Nunes da Rosa
- Center of Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas, Pelotas, RS, CEP: 96010-900, Brazil
| | - Isabel Ladeira Pereira
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, CEP: 96010-900, Brazil
- Biotechnology Unit, Technology Development Center, Federal University of Pelotas, Pelotas, RS, CEP: 96010-900, Brazil
| | | | - Daiane Drawanz Hartwig
- Department of Microbiology and Parasitology, Institute of Biology, Federal University of Pelotas, Pelotas, RS, CEP: 96010-900, Brazil.
- Biotechnology Unit, Technology Development Center, Federal University of Pelotas, Pelotas, RS, CEP: 96010-900, Brazil.
| |
Collapse
|
23
|
Nguyen HK, Picciotti SL, Duke MM, Broberg CA, Schoenfisch MH. Nitric Oxide-Induced Morphological Changes to Bacteria. ACS Infect Dis 2023; 9:2316-2324. [PMID: 37831756 PMCID: PMC11041245 DOI: 10.1021/acsinfecdis.3c00415] [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] [Indexed: 10/15/2023]
Abstract
Antimicrobial resistance poses a serious threat to global health, necessitating research for alternative approaches to treating infections. Nitric oxide (NO) is an endogenously produced molecule involved in multiple physiological processes, including the response to pathogens. Herein, we employed microscopy- and fluorescence-based techniques to investigate the effects of NO delivered from exogenous NO donors on the bacterial cell envelopes of pathogens, including resistant strains. Our goal was to assess the role of NO donor architecture (small molecules, oligosaccharides, dendrimers) on bacterial wall degradation to representative Gram-negative bacteria (Klebsiella pneumoniae, Pseudomonas aeruginosa) and Gram-positive bacteria (Staphylococcus aureus, Enterococcus faecium) upon treatment. Depending on the NO donor, bactericidal NO doses spanned 1.5-5.5 mM (total NO released). Transmission electron microscopy of bacteria following NO exposure indicated extensive membrane damage to Gram-negative bacteria with warping of the cellular shape and disruption of the cell wall. Among the small-molecule NO donors, those providing a more extended release (t1/2 = 120 min) resulted in greater damage to Gram-negative bacteria. In contrast, rapid NO release (t1/2 = 24 min) altered neither the morphology nor the roughness of these bacteria. For Gram-positive bacteria, NO treatments did not result in any drastic change to cellular shape or membrane integrity, despite permeation of the cell wall as measured by depolarization assays. The use of positively charged quaternary ammonium (QA)-modified NO-releasing dendrimer proved to be the only NO donor system capable of penetrating the thick peptidoglycan layer of Gram-positive bacteria.
Collapse
Affiliation(s)
- Huan K. Nguyen
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599
| | | | - Magdalena M. Duke
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599
| | | | - Mark H. Schoenfisch
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599
| |
Collapse
|
24
|
Noreika A, Stankevičiūtė J, Rutkienė R, Meškys R, Kalinienė L. Exploring the enzymatic activity of depolymerase gp531 from Klebsiella pneumoniae jumbo phage RaK2. Virus Res 2023; 336:199225. [PMID: 37741345 PMCID: PMC10550766 DOI: 10.1016/j.virusres.2023.199225] [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: 07/19/2023] [Revised: 08/28/2023] [Accepted: 09/14/2023] [Indexed: 09/25/2023]
Abstract
Klebsiella pneumoniae poses a major global challenge due to its virulence, multidrug resistance, and nosocomial nature. Thus, bacteriophage-derived proteins are extensively being investigated as a means to combat this bacterium. In this study, we explored the enzymatic specificity of depolymerase gp531, encoded by the jumbo bacteriophage vB_KleM_RaK2 (RaK2). We used two different methods to modify the reducing end of the oligosaccharides released during capsule hydrolysis with gp531. Subsequent acidic cleavage with TFA, followed by TLC and HPLC-MS analyses, revealed that RaK2 gp531 is a β-(1→4)-endoglucosidase. The enzyme specifically recognizes and cleaves the capsular polysaccharide (CPS) of the Klebsiella pneumoniae K54 serotype, releasing K-unit monomers (the main product), dimers, and trimers. Depolymerase gp531 remains active from 10 to 50 °C and in the pH 3-8 range, indicating its stability and versatility. Additionally, we demonstrated that gp531's activity is not affected by CPS acetylation, which is influenced by the growth conditions of the bacterial culture. Overall, our findings provide valuable insights into the enzymatic activity of the first characterized depolymerase targeting the capsule of the clinically relevant K54 serotype of K. pneumoniae.
Collapse
Affiliation(s)
- Algirdas Noreika
- Department of Molecular Microbiology and Biotechnology, Institute of Biochemistry, Life Sciences Center, Vilnius, Lithuania.
| | - Jonita Stankevičiūtė
- Department of Molecular Microbiology and Biotechnology, Institute of Biochemistry, Life Sciences Center, Vilnius, Lithuania
| | - Rasa Rutkienė
- Department of Molecular Microbiology and Biotechnology, Institute of Biochemistry, Life Sciences Center, Vilnius, Lithuania
| | - Rolandas Meškys
- Department of Molecular Microbiology and Biotechnology, Institute of Biochemistry, Life Sciences Center, Vilnius, Lithuania
| | - Laura Kalinienė
- Department of Molecular Microbiology and Biotechnology, Institute of Biochemistry, Life Sciences Center, Vilnius, Lithuania.
| |
Collapse
|
25
|
Wantuch PL, Rosen DA. Klebsiella pneumoniae: adaptive immune landscapes and vaccine horizons. Trends Immunol 2023; 44:826-844. [PMID: 37704549 DOI: 10.1016/j.it.2023.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 09/15/2023]
Abstract
Klebsiella pneumoniae is among the most common antibiotic-resistant pathogens causing nosocomial infections. Additionally, it is a leading cause of neonatal sepsis and childhood mortality across the globe. Despite its clinical importance, we are only beginning to understand how the mammalian adaptive immune system responds to this pathogen. Further, many studies investigating potential K. pneumoniae vaccine candidates or alternative therapies have been launched in recent years. Here, we review the current state of knowledge on the adaptive immune response to K. pneumoniae infections and progress towards developing vaccines and other therapies to combat these infections.
Collapse
Affiliation(s)
- Paeton L Wantuch
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David A Rosen
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
26
|
Kumar L, Bisen M, Harjai K, Chhibber S, Azizov S, Lalhlenmawia H, Kumar D. Advances in Nanotechnology for Biofilm Inhibition. ACS OMEGA 2023; 8:21391-21409. [PMID: 37360468 PMCID: PMC10286099 DOI: 10.1021/acsomega.3c02239] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023]
Abstract
Biofilm-associated infections have emerged as a significant public health challenge due to their persistent nature and increased resistance to conventional treatment methods. The indiscriminate usage of antibiotics has made us susceptible to a range of multidrug-resistant pathogens. These pathogens show reduced susceptibility to antibiotics and increased intracellular survival. However, current methods for treating biofilms, such as smart materials and targeted drug delivery systems, have not been found effective in preventing biofilm formation. To address this challenge, nanotechnology has provided innovative solutions for preventing and treating biofilm formation by clinically relevant pathogens. Recent advances in nanotechnological strategies, including metallic nanoparticles, functionalized metallic nanoparticles, dendrimers, polymeric nanoparticles, cyclodextrin-based delivery, solid lipid nanoparticles, polymer drug conjugates, and liposomes, may provide valuable technological solutions against infectious diseases. Therefore, it is imperative to conduct a comprehensive review to summarize the recent advancements and limitations of advanced nanotechnologies. The present Review encompasses a summary of infectious agents, the mechanisms that lead to biofilm formation, and the impact of pathogens on human health. In a nutshell, this Review offers a comprehensive survey of the advanced nanotechnological solutions for managing infections. A detailed presentation has been made as to how these strategies may improve biofilm control and prevent infections. The key objective of this Review is to summarize the mechanisms, applications, and prospects of advanced nanotechnologies to provide a better understanding of their impact on biofilm formation by clinically relevant pathogens.
Collapse
Affiliation(s)
- Lokender Kumar
- School
of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229, India
- Cancer
Biology Laboratory, Raj Khosla Centre for Cancer Research, Shoolini University, Solan, Himachal Pradesh 173229, India
| | - Monish Bisen
- School
of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229, India
| | - Kusum Harjai
- Department
of Microbiology, Panjab University, Chandigarh 160014, India
| | - Sanjay Chhibber
- Department
of Microbiology, Panjab University, Chandigarh 160014, India
| | - Shavkatjon Azizov
- Laboratory
of Biological Active Macromolecular Systems, Institute of Bioorganic
Chemistry, Academy of Sciences Uzbekistan, Tashkent 100125, Uzbekistan
- Faculty
of Life Sciences, Pharmaceutical Technical
University, Tashkent 100084, Uzbekistan
| | - Hauzel Lalhlenmawia
- Department
of Pharmacy, Regional Institute of Paramedical
and Nursing Sciences, Zemabawk, Aizawl, Mizoram 796017, India
| | - Deepak Kumar
- Department
of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh173229, India
| |
Collapse
|
27
|
Tsai CC, Lin JC, Chen PC, Liu EYM, Tsai YK, Yu CP, Li JJ, Wang CH, Fung CP, Lin FM, Chang FY, Siu LK. A 20-Year Study of Capsular Polysaccharide Seroepidemiology, Susceptibility Profiles, and Virulence Determinants of Klebsiella pneumoniae from Bacteremia Patients in Taiwan. Microbiol Spectr 2023; 11:e0035923. [PMID: 37191538 PMCID: PMC10269490 DOI: 10.1128/spectrum.00359-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/21/2023] [Indexed: 05/17/2023] Open
Abstract
In this study, we selected bacteremic Klebsiella pneumoniae isolates from the Taiwan Surveillance of Antimicrobial Resistance program. A total of 521 isolates were collected over a period of 2 decades, including 121 from 1998, 197 from 2008, and 203 from 2018. Seroepidemiology showed that the top five capsular polysaccharide types were serotypes K1, K2, K20, K54, and K62, constituting 48.5% of the total isolates, and the respective ratios at each time point have remained similar over the past 2 decades. The antibacterial susceptibility tests showed that K1, K2, K20, and K54 were susceptible to most antibiotics, while K62 was relatively resistant compared to other typeable and nontypeable strains. In addition, six virulence-associated genes, clbA, entB, iroN, rmpA, iutA, and iucA, were predominant in K1 and K2 isolates of K. pneumoniae. In conclusion, serotypes K1, K2, K20, K54, and K62 of K. pneumoniae are the most prevalent serotypes and carry more virulence determinants in bacteremia patients, which may indicate their invasiveness. If further serotype-specific vaccine development is performed, these five serotypes should be included. Since the antibiotic susceptibility profiles were stable over a long duration, empirical treatment may be predicted according to serotype if rapid diagnosis from direct clinical specimens is available, such as PCR or antigen serotyping for serotype K1 and K2. IMPORTANCE This is the first nationwide study to examine the seroepidemiology of Klebsiella pneumoniae using blood culture isolates collected over a period of 20 years. The study found that the prevalence of serotypes remained consistent over the 20-year period, with high-prevalence serotypes associated with invasive types. Nontypeable isolates had fewer virulence determinants than other serotypes. With the exception of serotype K62, the other high-prevalence serotypes were highly susceptible to antibiotics. If rapid diagnosis using direct clinical specimens, such as PCR or antigen serotyping, is available, empirical treatment can be predicted based on serotype, particularly for K1 and K2. The results of this seroepidemiology study could also help the development of future capsule polysaccharide vaccines.
Collapse
Affiliation(s)
- Chun-Chou Tsai
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jung-Chung Lin
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Pei-Chen Chen
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Esther Yip-Mei Liu
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Kuo Tsai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chia-Peng Yu
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Jia-Je Li
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ching-Hsun Wang
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chang-Phone Fung
- Section of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Fu-Mei Lin
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Feng-Yee Chang
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - L. Kristopher Siu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
28
|
Lazar V, Oprea E, Ditu LM. Resistance, Tolerance, Virulence and Bacterial Pathogen Fitness-Current State and Envisioned Solutions for the Near Future. Pathogens 2023; 12:pathogens12050746. [PMID: 37242416 DOI: 10.3390/pathogens12050746] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
The current antibiotic crisis and the global phenomena of bacterial resistance, inherited and non-inherited, and tolerance-associated with biofilm formation-are prompting dire predictions of a post-antibiotic era in the near future. These predictions refer to increases in morbidity and mortality rates as a consequence of infections with multidrug-resistant or pandrug-resistant microbial strains. In this context, we aimed to highlight the current status of the antibiotic resistance phenomenon and the significance of bacterial virulence properties/fitness for human health and to review the main strategies alternative or complementary to antibiotic therapy, some of them being already clinically applied or in clinical trials, others only foreseen and in the research phase.
Collapse
Affiliation(s)
- Veronica Lazar
- Department of Botany and Microbiology, Faculty of Biology, University of Bucharest, 1-3 Portocalelor Street, 060101 Bucharest, Romania
| | - Eliza Oprea
- Department of Botany and Microbiology, Faculty of Biology, University of Bucharest, 1-3 Portocalelor Street, 060101 Bucharest, Romania
| | - Lia-Mara Ditu
- Department of Botany and Microbiology, Faculty of Biology, University of Bucharest, 1-3 Portocalelor Street, 060101 Bucharest, Romania
| |
Collapse
|
29
|
Chen D, Srivastava AK, Dubrochowska J, Liu L, Li T, Hoffmann JP, Kolls JK, Boons GJ. A Bioactive Synthetic Outer-Core Oligosaccharide Derived from a Klebsiella pneumonia Lipopolysaccharide for Bacteria Recognition. Chemistry 2023; 29:e202203408. [PMID: 36662447 PMCID: PMC10159924 DOI: 10.1002/chem.202203408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/21/2023]
Abstract
There is an urgent need for new treatment options for carbapenem-resistant Klebsiella pneumoniae (K. pneumoniae), which is a common cause of life-threatening hospital- and community-acquired infections. Prophylactic or therapeutic vaccination may offer an approach to control these infections, however, none has yet been approved for human use. Here, we report the chemical synthesis of an outer core tetra- and pentasaccharide derived from the lipopolysaccharide of K. pneumoniae. The oligosaccharides were equipped with an aminopentyl linker, which facilitated conjugation to the carrier proteins CRM197 and BSA. Mice immunized with the glycoconjugate vaccine candidates elicited antibodies that recognized isolated LPS as well as various strains of K. pneumoniae. The successful preparation of the oligosaccharides relied on the selection of monosaccharide building blocks equipped with orthogonal hydroxyl and amino protecting groups. It allowed the differentiation of three types of amines of the target compounds and the installation of a crowded 4,5-branched Kdo moiety.
Collapse
Affiliation(s)
- Dushen Chen
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Akhilesh K Srivastava
- Department of Medicine and Pediatrics, Tulane School of Medicine, New Orleans, LA, USA
| | - Justyna Dubrochowska
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Lin Liu
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Tiehai Li
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | - Joseph P Hoffmann
- Department of Medicine and Pediatrics, Tulane School of Medicine, New Orleans, LA, USA
| | - Jay K Kolls
- Department of Medicine and Pediatrics, Tulane School of Medicine, New Orleans, LA, USA
| | - Geert-Jan Boons
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
- Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
- Chemistry Department, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
30
|
Wantuch PL, Knoot CJ, Robinson LS, Vinogradov E, Scott NE, Harding CM, Rosen DA. Capsular polysaccharide inhibits vaccine-induced O-antigen antibody binding and function across both classical and hypervirulent K2:O1 strains of Klebsiella pneumoniae. PLoS Pathog 2023; 19:e1011367. [PMID: 37146068 PMCID: PMC10191323 DOI: 10.1371/journal.ppat.1011367] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/17/2023] [Accepted: 04/17/2023] [Indexed: 05/07/2023] Open
Abstract
Klebsiella pneumoniae presents as two circulating pathotypes: classical K. pneumoniae (cKp) and hypervirulent K. pneumoniae (hvKp). Classical isolates are considered urgent threats due to their antibiotic resistance profiles, while hvKp isolates have historically been antibiotic susceptible. Recently, however, increased rates of antibiotic resistance have been observed in both hvKp and cKp, further underscoring the need for preventive and effective immunotherapies. Two distinct surface polysaccharides have gained traction as vaccine candidates against K. pneumoniae: capsular polysaccharide and the O-antigen of lipopolysaccharide. While both targets have practical advantages and disadvantages, it remains unclear which of these antigens included in a vaccine would provide superior protection against matched K. pneumoniae strains. Here, we report the production of two bioconjugate vaccines, one targeting the K2 capsular serotype and the other targeting the O1 O-antigen. Using murine models, we investigated whether these vaccines induced specific antibody responses that recognize K2:O1 K. pneumoniae strains. While each vaccine was immunogenic in mice, both cKp and hvKp strains exhibited decreased O-antibody binding in the presence of capsule. Further, O1 antibodies demonstrated decreased killing in serum bactericidal assays with encapsulated strains, suggesting that the presence of K. pneumoniae capsule blocks O1-antibody binding and function. Finally, the K2 vaccine outperformed the O1 vaccine against both cKp and hvKp in two different murine infection models. These data suggest that capsule-based vaccines may be superior to O-antigen vaccines for targeting hvKp and some cKp strains, due to capsule blocking the O-antigen.
Collapse
Affiliation(s)
- Paeton L. Wantuch
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Cory J. Knoot
- Omniose, Saint Louis, Missouri, United States of America
| | | | - Evgeny Vinogradov
- National Research Council Canada, Human Health Therapeutics Centre, Ottawa, Ontario, Canada
| | - Nichollas E. Scott
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | | | - David A. Rosen
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| |
Collapse
|
31
|
Martin MJ, Stribling W, Ong AC, Maybank R, Kwak YI, Rosado-Mendez JA, Preston LN, Lane KF, Julius M, Jones AR, Hinkle M, Waterman PE, Lesho EP, Lebreton F, Bennett JW, Mc Gann PT. A panel of diverse Klebsiella pneumoniae clinical isolates for research and development. Microb Genom 2023; 9:mgen000967. [PMID: 37141116 PMCID: PMC10272860 DOI: 10.1099/mgen.0.000967] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 01/27/2023] [Indexed: 05/05/2023] Open
Abstract
Klebsiella pneumoniae are a leading cause of healthcare-associated infections worldwide. In particular, strains expressing extended-spectrum β-lactamases (ESBLs) and carbapenemases pose serious treatment challenges, leading the World Health Organization (WHO) to designate ESBL and carbapenem-resistant Enterobacteriaceae as 'critical' threats to human health. Research efforts to combat these pathogens can be supported by accessibility to diverse and clinically relevant isolates for testing novel therapeutics. Here, we describe a panel of 100 diverse K. pneumoniae isolates that are publicly available to assist the research community in this endeavour. Whole-genome sequencing (WGS) was performed on 3878 K. pneumoniae clinical isolates housed at the Multidrug-Resistant Organism Repository and Surveillance Network. The isolates were cultured from 63 facilities in 19 countries between 2001 and 2020. Core-genome multilocus sequence typing and high-resolution single-nucleotide polymorphism-based phylogenetic analyses captured the genetic diversity of the collection and were used to select the final panel of 100 isolates. In addition to known multidrug-resistant (MDR) pandemic lineages, the final panel includes hypervirulent lineages and isolates with specific and diverse resistance genes and virulence biomarkers. A broad range of antibiotic susceptibilities, ranging from pan-sensitive to extensively drug-resistant isolates, are described. The panel collection, and all associated metadata and genome sequences, are available at no additional cost and will be an important resource for the research community and for the design and development of novel antimicrobial agents and diagnostics against this important pathogen.
Collapse
Affiliation(s)
- Melissa J. Martin
- Multidrug-Resistant Organism Repository and Surveillance Network, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - William Stribling
- Multidrug-Resistant Organism Repository and Surveillance Network, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Ana C. Ong
- Multidrug-Resistant Organism Repository and Surveillance Network, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Rosslyn Maybank
- Multidrug-Resistant Organism Repository and Surveillance Network, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Yoon I. Kwak
- Multidrug-Resistant Organism Repository and Surveillance Network, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Joshua A. Rosado-Mendez
- Multidrug-Resistant Organism Repository and Surveillance Network, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Lan N. Preston
- Multidrug-Resistant Organism Repository and Surveillance Network, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Katharine F. Lane
- Multidrug-Resistant Organism Repository and Surveillance Network, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Michael Julius
- Multidrug-Resistant Organism Repository and Surveillance Network, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Anthony R. Jones
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Mary Hinkle
- Infectious Diseases Unit, Rochester General Hospital, Rochester, New York, USA
| | - Paige E. Waterman
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Emil P. Lesho
- Infectious Diseases Unit, Rochester General Hospital, Rochester, New York, USA
| | - Francois Lebreton
- Multidrug-Resistant Organism Repository and Surveillance Network, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Jason W. Bennett
- Multidrug-Resistant Organism Repository and Surveillance Network, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Patrick T. Mc Gann
- Multidrug-Resistant Organism Repository and Surveillance Network, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| |
Collapse
|
32
|
St. John A, Perault AI, Giacometti SI, Sommerfield AG, DuMont AL, Lacey KA, Zheng X, Sproch J, Petzold C, Dancel-Manning K, Gonzalez S, Annavajhala M, Beckford C, Zeitouni N, Liang FX, van Bakel H, Shopsin B, Uhlemann AC, Pironti A, Torres VJ. Capsular Polysaccharide Is Essential for the Virulence of the Antimicrobial-Resistant Pathogen Enterobacter hormaechei. mBio 2023; 14:e0259022. [PMID: 36779722 PMCID: PMC10127600 DOI: 10.1128/mbio.02590-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/13/2023] [Indexed: 02/14/2023] Open
Abstract
Nosocomial infections caused by multidrug-resistant (MDR) Enterobacter cloacae complex (ECC) pathogens are on the rise. However, the virulence strategies employed by these pathogens remain elusive. Here, we study the interaction of ECC clinical isolates with human serum to define how this pathogen evades the antimicrobial action of complement, one of the first lines of host-mediated immune defense. We identified a small number of serum-sensitive strains, including Enterobacter hormaechei strain NR3055, which we exploited for the in vitro selection of serum-resistant clones. Comparative genomics between the serum-sensitive NR3055 strain and the isolated serum-resistant clones revealed a premature stop codon in the wzy gene of the capsular polysaccharide biosynthesis locus of NR3055. The complementation of wzy conferred serum resistance to NR3055, prevented the deposition of complement proteins on the bacterial surface, inhibited phagocytosis by human neutrophils, and rendered the bacteria virulent in a mouse model of peritonitis. Mice exposed to a nonlethal dose of encapsulated NR3055 were protected from subsequent lethal infections by encapsulated NR3055, whereas mice that were previously exposed to unencapsulated NR3055 succumbed to infection. Thus, capsule is a key immune evasion determinant for E. hormaechei, and it is a potential target for prophylactics and therapeutics to combat these increasingly MDR human pathogens. IMPORTANCE Infections caused by antimicrobial resistant bacteria are of increasing concern, especially those due to carbapenem-resistant Enterobacteriaceae pathogens. Included in this group are species of the Enterobacter cloacae complex, regarding which there is a paucity of knowledge on the infection biology of the pathogens, despite their clinical relevance. In this study, we combine techniques in comparative genomics, bacterial genetics, and diverse models of infection to establish capsule as an important mechanism of Enterobacter pathogens to resist the antibacterial activity of serum, a first line of host defense against bacterial infections. We also show that immune memory targeting the Enterobacter capsule protects against lethal infection. The further characterization of Enterobacter infection biology and the immune response to infection are needed for the development of therapies and preventative interventions targeting these highly antibiotic resistant pathogens.
Collapse
Affiliation(s)
- Amelia St. John
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
| | - Andrew I. Perault
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
| | - Sabrina I. Giacometti
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| | - Alexis G. Sommerfield
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Ashley L. DuMont
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Keenan A. Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Xuhui Zheng
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Julia Sproch
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Chris Petzold
- Microscopy Laboratory, Division of Advanced Research Technologies, New York University Langone Health, New York, New York, USA
| | - Kristen Dancel-Manning
- Microscopy Laboratory, Division of Advanced Research Technologies, New York University Langone Health, New York, New York, USA
| | - Sandra Gonzalez
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Medini Annavajhala
- Department of Medicine, Division of Infectious Diseases, Columbia University Medical Center, New York, New York, USA
| | - Colleen Beckford
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nathalie Zeitouni
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Feng-Xia Liang
- Microscopy Laboratory, Division of Advanced Research Technologies, New York University Langone Health, New York, New York, USA
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, New York, USA
| | - Anne-Catrin Uhlemann
- Department of Medicine, Division of Infectious Diseases, Columbia University Medical Center, New York, New York, USA
| | - Alejandro Pironti
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Microbial Computational Genomic Core Lab, Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
33
|
Wang W, Cao Y, Li J, Lu S, Ge H, Pan S, Pan X, Wang L. The impact of osmotic stresses on the biofilm formation, immunodetection, and morphology of Aeromonas hydrophila. Microbiol Res 2023; 269:127301. [PMID: 36689842 DOI: 10.1016/j.micres.2023.127301] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/29/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
Aeromonas hydrophila (Ah) is a zoonotic pathogen of great importance to aquaculture and human health. This study systematically evaluated the impact of salinity, sugar, ammonia nitrogen, and nitric nitrogen levels on the fitness of Ah by using Luria-Bertani (LB) broth supplemented with different concentrations of NaCl, sucrose, NH4Cl, urea, NaNO2 or NaNO3. Results showed that the static biofilm formation of Ah was higher at 28 °C compared to 37 °C (P < 0.05). At 28 °C, as the NaCl (>1 %) and sucrose levels increased, the Ah biofilm formation and the binding between Ah cells and monoclonal antibodies (mAbs, for immunodetection) decreased. Elevated ammonia nitrogen and nitric nitrogen levels generated no significant impact on Ah biofilm formation or immunodetection (P > 0.05). The expression of mAbs-targeted Omp remained unchanged under high NaCl or sucrose conditions. Further analysis showed that high sucrose conditions led to the over-expression of the extracellular polysaccharides (PS) and promoted the formation of capsule-like structures. These over-expressed PS and capsule structures might be one reason explaining the inhibited immunodetection efficacy. Results generated from this study provide crucial insights for the design of recovery and detection protocols for Ah present in food or environmental samples.
Collapse
Affiliation(s)
- Wenbin Wang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, Jiangsu, China; Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture and Rural Affairs; Key Laboratory of Fish Health and Nutrition of Zhejiang Province; Zhejiang Institute of Freshwater Fisheries, Huzhou, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, Jiangsu, China; Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Ye Cao
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, Jiangsu, China; Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Jing Li
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, Jiangsu, China; Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Shuaichen Lu
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, Jiangsu, China; Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Hongxing Ge
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, Jiangsu, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, Jiangsu, China; Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Saikun Pan
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, Jiangsu, China; Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture and Rural Affairs; Key Laboratory of Fish Health and Nutrition of Zhejiang Province; Zhejiang Institute of Freshwater Fisheries, Huzhou, China; Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang, Jiangsu, China; Jiangsu Key Laboratory of Marine Biotechnology, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Xiaoyi Pan
- Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture and Rural Affairs; Key Laboratory of Fish Health and Nutrition of Zhejiang Province; Zhejiang Institute of Freshwater Fisheries, Huzhou, China.
| | - Luxin Wang
- Department of Food Science and Technology, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
34
|
Gautam I, Huss CW, Storad ZA, Krebs M, Bassiouni O, Ramesh R, Wuescher LM, Worth RG. Activated Platelets Mediate Monocyte Killing of Klebsiella pneumoniae. Infect Immun 2023; 91:e0055622. [PMID: 36853027 PMCID: PMC10016073 DOI: 10.1128/iai.00556-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/02/2023] [Indexed: 03/01/2023] Open
Abstract
Platelets are known for essential activities in hemostasis and for their important contribution to protection against infectious pathogens. Klebsiella pneumoniae is an opportunistic pathogen widely known to cause nosocomial infections. Recently, hypervirulent strains of K. pneumoniae have been emerging, which can cause severe infections in immunocompetent individuals. Combined with the increase in antibiotic resistance, it is important to understand how K. pneumoniae affects components of the immune system. We studied the interactions of human platelets with several K. pneumoniae strains (the wild type encapsulated strain, and a nonencapsulated mutant). Thrombin-stimulated whole human and mouse blood significantly inhibited bacterial growth compared to unstimulated whole blood. Furthermore, we investigated the effect of K. pneumoniae on platelet activation. Both strains induced significant increase in activation of both unstimulated and thrombin-stimulated human platelets. Additionally, only the nonencapsulated mutant increased aggregation of platelets in response to ADP. K. pneumoniae killing assays were then performed with washed platelets in the presence or absence of thrombin. Surprisingly, washed platelets failed to exhibit any effects on the growth of K. pneumoniae. We further explored the impact of platelets on monocyte-mediated killing of K. pneumoniae. Importantly, we found that activated platelets significantly enhanced monocyte-mediated killing of K. pneumoniae. This effect was likely due to the formation of platelet-monocyte aggregates in blood upon thrombin stimulation. Overall, this study highlights the role of platelets in mediating a protective response against K. pneumoniae and reinforces the importance of platelets in modulating leukocyte behavior.
Collapse
Affiliation(s)
- Iluja Gautam
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Chadwick W. Huss
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Zachary A. Storad
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Michelle Krebs
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Omar Bassiouni
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Rochan Ramesh
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Leah M. Wuescher
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Randall G. Worth
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| |
Collapse
|
35
|
Ashwath P, Somanath D, Sannejal AD. CRISPR and Antisense RNA Technology: Exploiting Nature's Tool to Restrain Virulence in Tenacious Pathogens. Mol Biotechnol 2023; 65:17-27. [PMID: 35980592 DOI: 10.1007/s12033-022-00539-4] [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: 04/11/2022] [Accepted: 07/25/2022] [Indexed: 01/11/2023]
Abstract
Pathogenic bacteria constitute a significant threat to mankind and at the same time represent a huge reservoir of abeyant therapeutics to prevent and treat various diseases. The concept of virulence determinants has been a compelling tool in driving research in the field of bacterial pathogenesis and infectious diseases. In this review, we highlight a few virulence elements forged by the pathogens from the viewpoint of the damage-response scaffold, vandalizing the susceptible host. Seeking an alternative to target the virulence determinants heads a road map toward the exemplary molecular approach. Hence, here we explore some of the exceptional applications of the clustered regulatory interspaced short palindromic repeat (CRISPR)- based therapy and antisense RNA (asRNA) approach, which could be exploited to selectively dismantle adamant components of the pathogen's virulence machinery. To the best of our knowledge, this is the first review paper involving both CRISPR and antisense RNA technology, as an alternative strategy to evade virulence mechanisms in bacterial pathogens.
Collapse
Affiliation(s)
- Priyanka Ashwath
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research, Deralakatte, Mangaluru, 575018, India
| | - Disha Somanath
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research, Deralakatte, Mangaluru, 575018, India
| | - Akhila Dharnappa Sannejal
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research, Deralakatte, Mangaluru, 575018, India.
| |
Collapse
|
36
|
Liang Z, Wang Y, Lai Y, Zhang J, Yin L, Yu X, Zhou Y, Li X, Song Y. Host defense against the infection of Klebsiella pneumoniae: New strategy to kill the bacterium in the era of antibiotics? Front Cell Infect Microbiol 2022; 12:1050396. [PMID: 36506034 PMCID: PMC9730340 DOI: 10.3389/fcimb.2022.1050396] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/10/2022] [Indexed: 11/25/2022] Open
Abstract
Klebsiella pneumoniae (K. pneumoniae) is a typical gram-negative iatrogenic bacterium that often causes bacteremia, pneumonia and urinary tract infection particularly among those with low immunity. Although antibiotics is the cornerstone of anti-infections, the clinical efficacy of β-lactamase and carbapenems drugs has been weakened due to the emergence of drug-resistant K. pneumoniae. Recent studies have demonstrated that host defense plays a critical role in killing K. pneumoniae. Here, we summarize our current understanding of host immunity mechanisms against K. pneumoniae, including mechanical barrier, innate immune cells, cellular immunity and humoral immunity, providing a theoretical basis and the new strategy for the clinical treatment of K. pneumoniae through improving host immunity.
Collapse
Affiliation(s)
- Zihan Liang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Yiyao Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Yixiang Lai
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Jingyi Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Lanlan Yin
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Xiang Yu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Yongqin Zhou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Xinzhi Li
- College of Basic Medical Science, China Three Gorges University, Yichang, China,Affiliated Renhe Hospital of China Three Gorges University, Yichang, China,*Correspondence: Yinhong Song, ; Xinzhi Li,
| | - Yinhong Song
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China,*Correspondence: Yinhong Song, ; Xinzhi Li,
| |
Collapse
|
37
|
Identification and antibacterial evaluation of endophytic actinobacteria from Luffa cylindrica. Sci Rep 2022; 12:18236. [PMID: 36309579 PMCID: PMC9617871 DOI: 10.1038/s41598-022-23073-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 10/25/2022] [Indexed: 12/31/2022] Open
Abstract
The emergence of antibiotic-resistant bacteria has limited treatment options and led to the untreatable infections, thereby necessitating the discovery of new antibiotics to battel against bacteria. Natural products from endophytic actinobacteria (EA) serve as a reservoir for discovery of new antibiotics. Therefore, the current study focused on the isolation and antibacterial properties of EA isolated from Luffa cylindrica. Six strains were identified using morphological characterization, SEM analyses and 16S rRNA gene sequencing from the roots and leaves of the plant. They were taxonomically classified as Streptomycetaceae family. This is the first report on EA form L. cylindrica. The strains produced a chain of oval, cubed or cylindrical shaped spores with spiny or smooth surfaces. Three strains; KUMS-B3, KUMS-B4 and KUMS-B6 were reported as endophytes for the first time. Fifty percent of isolates were isolated from leaves samples using YECD medium. Our results showed that the sampling time and seasons may affect the bacterial diversity. All six strains had antibacterial activity against at least one of the tested bacteria S. aureus, P. aeruginosa, and E. coli. Among the strains, KUMS-B6 isolate, closely related to S. praecox, exhibited the highest antibacterial activity against both gram-positive and negative bacteria. KUMS-B6, KUMS-B5 and KUMS-B4 isolates strongly inhibited the growth of P. aeruginosa. Interestingly, the strains, isolated from leaves exhibited stronger antagonist activities compared to those isolated from the roots. The study revealed that the isolated strains from Luffa produce a plethora of bioactive substances that are potential source of new drug candidates for the treatment of infections.
Collapse
|
38
|
da Silva Junior AG, Frias IAM, Lima-Neto RG, Franco OL, Oliveira MDL, Andrade CAS. Electrochemical detection of gram-negative bacteria through mastoparan-capped magnetic nanoparticle. Enzyme Microb Technol 2022; 160:110088. [PMID: 35777193 DOI: 10.1016/j.enzmictec.2022.110088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/09/2022] [Accepted: 06/16/2022] [Indexed: 12/29/2022]
Abstract
The increasing number of multidrug resistance microorganisms is an alarming threat, and their rapid detection is essential to prevent nosocomial, foodborne, or waterborne infections. Many peptides derived from the venom of wasp Synoeca surinama have antimicrobial activity against Gram-positive and Gram-negative bacteria. Synoeca-MP, an antimicrobial peptide (AMP) from mastoparan family, seems to increase bacterial membrane permeability, promoting cytotoxicity and membrane disruption. Here Synoeca-MP was evaluated as biorecognition element tethered over chitosan-coated magnetic nanoparticles (Fe3O4-Chit). The transducing layer of the biosensor was developed from the self-assembling of 4-mercaptobenzoic acid (4-MBA) monolayer onto gold substrate. Atomic force microscopy (AFM) analyses confirmed the biointeraction between AMP and different pathogens membranes. The fabrication and performance of the biosensing assembly were characterized by cyclic voltammetry (CV) and electrochemical impedance spectroscopy (EIS). Detection of Enterococcus faecalis (G+), Klebsiella pneumoniae (G-), Pseudomonas aeruginosa (G-), and Candida tropicalis was assessed in a recognition range from 101 to 105 CFU.mL-1. An instrumental limit of detection of 10 CFU.mL-1 was obtained for each specimen. However, the device presented a preferential selectivity towards Gram-negative bacteria. The proposed biosensor is a sensitive, fast, and straightforward platform for microbial detection in aqueous samples, envisaged for environmental monitoring applications.
Collapse
Affiliation(s)
- Alberto G da Silva Junior
- Programa de Pós-Graduação em Inovação Terapêutica, Universidade Federal de Pernambuco, 50670-901 Recife, PE, Brazil; Laboratório de Biodispositivos Nanoestruturados, Departamento de Bioquímica, Universidade Federal de Pernambuco, 50670-901 Recife, PE, Brazil
| | - Isaac A M Frias
- Programa de Pós-Graduação em Inovação Terapêutica, Universidade Federal de Pernambuco, 50670-901 Recife, PE, Brazil; Laboratório de Biodispositivos Nanoestruturados, Departamento de Bioquímica, Universidade Federal de Pernambuco, 50670-901 Recife, PE, Brazil
| | - Reginaldo G Lima-Neto
- Centro de Ciências da Saúde, Departamento de Medicina Tropical, Universidade Federal de Pernambuco, 50670-901 Recife, PE, Brazil
| | - Octávio L Franco
- Centro de Análises Proteômicas e Bioquímicas de Brasília, Pos-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil; S-Inova Biotech, Pos-Graduação em Biotecnologia, Universidade Católica Dom Bosco, MS, Brazil
| | - Maria D L Oliveira
- Programa de Pós-Graduação em Inovação Terapêutica, Universidade Federal de Pernambuco, 50670-901 Recife, PE, Brazil; Laboratório de Biodispositivos Nanoestruturados, Departamento de Bioquímica, Universidade Federal de Pernambuco, 50670-901 Recife, PE, Brazil
| | - César A S Andrade
- Programa de Pós-Graduação em Inovação Terapêutica, Universidade Federal de Pernambuco, 50670-901 Recife, PE, Brazil; Laboratório de Biodispositivos Nanoestruturados, Departamento de Bioquímica, Universidade Federal de Pernambuco, 50670-901 Recife, PE, Brazil.
| |
Collapse
|
39
|
Banerjee K, Motley MP, Boniche-Alfaro C, Bhattacharya S, Shah R, Ardizzone A, Fries BC. Patient-Derived Antibody Data Yields Development of Broadly Cross-Protective Monoclonal Antibody against ST258 Carbapenem-Resistant Klebsiella pneumoniae. Microbiol Spectr 2022; 10:e0176022. [PMID: 35862974 PMCID: PMC9430753 DOI: 10.1128/spectrum.01760-22] [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: 06/06/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022] Open
Abstract
The most pressing challenge for the development of anti-capsular antibodies is maximizing coverage against the heterogenous capsular polysaccharide (CPS) of carbapenem-resistant Klebsiella pneumoniae (CR-Kp). So far, only CR-Kp with wzi154 CPS has been successfully targeted by antibodies. Here, we present murine antibody 24D11, which was developed by vaccinating mice with purified wzi50-type CPS. Cross-reactivity and protective efficacy of MAb 24D11 were confirmed against CR-Kp that express the 3 most prevalent CPS types (wzi29, wzi154, wzi50) using both in vitro and in vivo infection models. 24D11 induced complement-mediated and independent opsonophagocytosis in macrophages as well as killing of all CR-Kp strains in whole blood cells derived from healthy donors. In a murine intratracheal infection model, 24D11 reduced lung burden and dissemination of CR-Kp strains when administered 4 h pre- or postinfection. The protective efficacy of 24D11 remained effective in neutropenic mice. This is the first antibody which exhibits cross-protective efficacy against clade 1 and 2 ST258 CR-Kp strains. It overcomes a major barrier to successfully target wzi29, a major CPS expressed by ST258 CR-Kp. The finding that 24D11 also exhibits potent protective efficacy against wzi154 CR-Kp strains highlights its high potential as a lead agent for the development of broadly active immunotherapy. IMPORTANCE Here, we present in vitro and in vivo data for the wzi50 CPS-specific monoclonal antibody MAb 24D11, demonstrating its cross-protective efficacy against three prominent win types (wzi29, wzi154, and wzi50) of the carbapenem-resistant clonal group CG258. In a murine pulmonary infection model, MAb 24D11 reduced bacterial lung burden and dissemination to other organs even if administered 4 h postinfection. Its protective efficacy was also observed in neutropenic mice, which highlights its potential value in clinical settings where oncology patients with CG258 infections may also be neutropenic.
Collapse
Affiliation(s)
- Kasturi Banerjee
- Department of Medicine, Infectious Disease Division, Stony Brook University, Stony Brook, New York, USA
- Veteran’s Administration Medical Center, Northport, New York, USA
| | - Michael P. Motley
- Department of Medicine, Infectious Disease Division, Stony Brook University, Stony Brook, New York, USA
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| | - Camila Boniche-Alfaro
- Department of Medicine, Infectious Disease Division, Stony Brook University, Stony Brook, New York, USA
- Veteran’s Administration Medical Center, Northport, New York, USA
| | - Somanon Bhattacharya
- Department of Medicine, Infectious Disease Division, Stony Brook University, Stony Brook, New York, USA
| | - Raj Shah
- Department of Medicine, Infectious Disease Division, Stony Brook University, Stony Brook, New York, USA
| | - Andrew Ardizzone
- Department of Medicine, Infectious Disease Division, Stony Brook University, Stony Brook, New York, USA
| | - Bettina C. Fries
- Department of Medicine, Infectious Disease Division, Stony Brook University, Stony Brook, New York, USA
- Veteran’s Administration Medical Center, Northport, New York, USA
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
40
|
Garcia Mendez DF, Rengifo Herrera JA, Sanabria J, Wist J. Analysis of the Metabolic Response of Planktonic Cells and Biofilms of Klebsiella pneumoniae to Sublethal Disinfection with Sodium Hypochlorite Measured by NMR. Microorganisms 2022; 10:1323. [PMID: 35889041 PMCID: PMC9323045 DOI: 10.3390/microorganisms10071323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
Klebsiella pneumoniae is a pathogenic agent able to form biofilms on water storage tanks and pipe walls. This opportunistic pathogen can generate a thick layer as one of its essential virulence factors, enabling the bacteria to survive disinfection processes and thus develop drug resistance. Understanding the metabolic differences between biofilm and planktonic cells of the K. pneumoniae response to NaClO is key to developing strategies to control its spread. In this study, we performed an NMR metabolic profile analysis to compare the response to a sublethal concentration of sodium hypochlorite of biofilm and planktonic cells of K. pneumoniae cultured inside silicone tubing. Metabolic profiles revealed changes in the metabolism of planktonic cells after a contact time of 10 min with 7 mg L-1 of sodium hypochlorite. A decrease in the production of metabolites such as lactate, acetate, ethanol, and succinate in this cell type was observed, thus indicating a disruption of glucose intake. In contrast, the biofilms displayed a high metabolic heterogeneity, and the treatment did not affect their metabolic signature.
Collapse
Affiliation(s)
- David Felipe Garcia Mendez
- Chemistry Department, Universidad del Valle—Sede Meléndez, Cali 13 # 100-00, Colombia; (D.F.G.M.); (J.W.)
- Australian National Phenome Center, Murdoch University, Perth, WA 6150, Australia
| | - Julián Andrés Rengifo Herrera
- Centro de Investigación y Desarrollo en Ciencias Aplicadas “Dr. J.J. Ronco” (CINDECA), Departamento de Química, Facultad de Ciencias Exactas, UNLP-CCT La Plata, CONICET, 47 No. 257, La Plata 1900, Argentina;
| | - Janeth Sanabria
- Australian National Phenome Center, Murdoch University, Perth, WA 6150, Australia
- Environmental Microbiology and Biotechnology Laboratory, Engineering Faculty, Engineering School of Environmental & Natural Resources, Universidad del Valle—Meléndez Campus, Cali 13 # 100-00, Colombia
| | - Julien Wist
- Chemistry Department, Universidad del Valle—Sede Meléndez, Cali 13 # 100-00, Colombia; (D.F.G.M.); (J.W.)
- Australian National Phenome Center, Murdoch University, Perth, WA 6150, Australia
| |
Collapse
|
41
|
Candida Worsens Klebsiella pneumoniae Induced-Sepsis in a Mouse Model with Low Dose Dextran Sulfate Solution through Gut Dysbiosis and Enhanced Inflammation. Int J Mol Sci 2022; 23:ijms23137050. [PMID: 35806054 PMCID: PMC9266745 DOI: 10.3390/ijms23137050] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
Klebsiella pneumoniae is an opportunistic pathogen and a commensal organism that is possibly enhanced in several conditions with gut dysbiosis, and frequently detectable together with Candida overgrowth. Here, K. pneumoniae with or without Candida albicans was daily orally administered for 3 months in 0.8% dextran sulfate solution-induced mucositis mice and also tested in vitro. As such, Candida worsened Klebsiella-DSS-colitis as demonstrated by mortality, leaky gut (FITC-dextran assay, bacteremia, endotoxemia, and serum beta-glucan), gut dysbiosis (increased Deferribacteres from fecal microbiome analysis), liver pathology (histopathology), liver apoptosis (activated caspase 3), and cytokines (in serum and in the internal organs) when compared with Klebsiella-administered DSS mice. The combination of heat-killed Candida plus Klebsiella mildly facilitated inflammation in enterocytes (Caco-2), hepatocytes (HepG2), and THP-1-derived macrophages as indicated by supernatant cytokines or the gene expression. The addition of heat-killed Candida into Klebsiella preparations upregulated TLR-2, reduced Occludin (an intestinal tight junction molecule), and worsened enterocyte integrity (transepithelial electrical resistance) in Caco-2 and enhanced casp8 and casp9 (apoptosis genes) in HepG2 when compared with heat-killed Klebsiella alone. In conclusion, Candida enhanced enterocyte inflammation (partly through TLR-2 upregulation and gut dysbiosis) that induced gut translocation of endotoxin and beta-glucan causing hyper-inflammatory responses, especially in hepatocytes and macrophages.
Collapse
|
42
|
Hua Y, Wu Y, Guo M, Ma R, Li Q, Hu Z, Chen H, Zhang X, Li H, Li Q, He P. Characterization and Functional Studies of a Novel Depolymerase Against K19-Type Klebsiella pneumoniae. Front Microbiol 2022; 13:878800. [PMID: 35814656 PMCID: PMC9257171 DOI: 10.3389/fmicb.2022.878800] [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: 02/18/2022] [Accepted: 05/31/2022] [Indexed: 12/14/2022] Open
Abstract
Carbapenem-resistant Klebsiella pneumoniae (CRKP), a pathogen that causes severe nosocomial infections and yields a high mortality rate, poses a serious threat to global public health due to its high antimicrobial resistance. Bacteriophages encode polysaccharide-degrading enzymes referred to as depolymerases that cleave the capsular polysaccharide (CPS), one of the main virulence factors of K. pneumoniae. In this study, we identified and characterized a new capsule depolymerase K19-Dpo41 from K. pneumoniae bacteriophage SH-KP156570. Our characterization of K19-Dpo41 demonstrated that this depolymerase showed specific activities against K19-type K. pneumoniae. K19-Dpo41-mediated treatments promoted the sensitivity of a multidrug-resistant K19-type K. pneumoniae strain to the bactericidal effect of human serum and significantly increased the survival rate of Galleria mellonella infected with K19-type K. pneumoniae. Our results provided strong primary evidence that K19-Dpo41 was not only effective in capsular typing of K19-type K. pneumoniae but promising in terms of developing new alternative therapeutic strategies against K19-type CRKP infections in the future.
Collapse
Affiliation(s)
- Yunfen Hua
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongqin Wu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minjie Guo
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, United States
| | - Ruijing Ma
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
| | - Qingchuan Li
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheyuan Hu
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongrui Chen
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingyu Zhang
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Li
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingtian Li
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Qingtian Li,
| | - Ping He
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
- Ping He,
| |
Collapse
|
43
|
Urso A, Prince A. Anti-Inflammatory Metabolites in the Pathogenesis of Bacterial Infection. Front Cell Infect Microbiol 2022; 12:925746. [PMID: 35782110 PMCID: PMC9240774 DOI: 10.3389/fcimb.2022.925746] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/23/2022] [Indexed: 01/13/2023] Open
Abstract
Host and pathogen metabolism have a major impact on the outcome of infection. The microenvironment consisting of immune and stromal cells drives bacterial proliferation and adaptation, while also shaping the activity of the immune system. The abundant metabolites itaconate and adenosine are classified as anti-inflammatory, as they help to contain the local damage associated with inflammation, oxidants and proteases. A growing literature details the many roles of these immunometabolites in the pathogenesis of infection and their diverse functions in specific tissues. Some bacteria, notably P. aeruginosa, actively metabolize these compounds, others, such as S. aureus respond by altering their own metabolic programs selecting for optimal fitness. For most of the model systems studied to date, these immunometabolites promote a milieu of tolerance, limiting local immune clearance mechanisms, along with promoting bacterial adaptation. The generation of metabolites such as adenosine and itaconate can be host protective. In the setting of acute inflammation, these compounds also represent potential therapeutic targets to prevent infection.
Collapse
Affiliation(s)
| | - Alice Prince
- *Correspondence: Alice Prince, ; Andreacarola Urso,
| |
Collapse
|
44
|
Assefa M. Multi-drug resistant gram-negative bacterial pneumonia: etiology, risk factors, and drug resistance patterns. Pneumonia (Nathan) 2022; 14:4. [PMID: 35509063 PMCID: PMC9069761 DOI: 10.1186/s41479-022-00096-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/11/2022] [Indexed: 12/27/2022] Open
Abstract
Bacterial pneumonia is one of the most serious public health issues owing to its medical and economic costs, which result in increased morbidity and mortality in people of all ages around the world. Furthermore, antimicrobial resistance has risen over time, and the advent of multi-drug resistance in GNB complicates therapy and has a detrimental impact on patient outcomes. The current review aimed to summarize bacterial pneumonia with an emphasis on gram-negative etiology, pathogenesis, risk factors, resistance mechanisms, treatment updates, and vaccine concerns to tackle the problem before it causes a serious consequence. In conclusion, the global prevalence of GNB in CAP was reported 49.7% to 83.1%, whereas in VAP patients ranged between 76.13% to 95.3%. The most commonly reported MDR-GNB causes of pneumonia were A. baumannii, K. pneumoniae, and P. aeruginosa, with A. baumannii isolated particularly in VAP patients and the elderly. In most studies, ampicillin, tetracyclines, amoxicillin-clavulanic acid, cephalosporins, and carbapenems were shown to be highly resistant. Prior MDR-GNB infection, older age, previous use of broad-spectrum antibiotics, high frequency of local antibiotic resistance, prolonged hospital stays, ICU admission, mechanical ventilation, and immunosuppression are associated with the MDR-GNB colonization. S. maltophilia was reported as a severe cause of HAP/VAP in patients with mechanically ventilated and having hematologic malignancy due to its ability of biofilm formation, site adhesion in respiratory devices, and its intrinsic and acquired drug resistance mechanisms. Effective combination therapies targeting PDR strains and drug-resistant genes, antibiofilm agents, gene-based vaccinations, and pathogen-specific lymphocytes should be developed in the future.
Collapse
Affiliation(s)
- Muluneh Assefa
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, P.O. Box 196, Gondar, Ethiopia.
| |
Collapse
|
45
|
Xu C, Dong N, Chen K, Yang X, Zeng P, Hou C, Chi Chan EW, Yao X, Chen S. Bactericidal, anti-biofilm, and anti-virulence activity of vitamin C against carbapenem-resistant hypervirulent Klebsiella pneumoniae. iScience 2022; 25:103894. [PMID: 35243252 PMCID: PMC8873610 DOI: 10.1016/j.isci.2022.103894] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/15/2021] [Accepted: 02/04/2022] [Indexed: 12/27/2022] Open
Affiliation(s)
- Chen Xu
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Ning Dong
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Kaichao Chen
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Xuemei Yang
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Ping Zeng
- State Key Lab of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Changshun Hou
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Edward Wai Chi Chan
- State Key Lab of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Xi Yao
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Sheng Chen
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Corresponding author
| |
Collapse
|
46
|
Dey J, Mahapatra SR, Lata S, Patro S, Misra N, Suar M. Exploring Klebsiella pneumoniae capsule polysaccharide proteins to design multiepitope subunit vaccine to fight against pneumonia. Expert Rev Vaccines 2022; 21:569-587. [PMID: 34932430 DOI: 10.1080/14760584.2022.2021882] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Klebsiella pneumoniae is an emerging human pathogen causing neonatal lung disease, catheter-associated infections, and nosocomial outbreaks with high fatality rates. Capsular polysaccharide (CPS) protein plays a major determinant in virulence and is considered as a promising target for vaccine development. RESEARCH DESIGN AND METHODS In this study, we used immunoinformatic approaches to design a multi-peptide vaccine against K. pneumonia. The epitopes were selected through several immune filters, such as antigenicity, conservancy, nontoxicity, non-allergenicity, binding affinity to HLA alleles, overlapping epitopes, and peptides having common epitopes. RESULTS Finally, a construct comprising 2 B-Cell, 8 CTL, 2 HTL epitopes, along with adjuvant, linkers was designed. Peptide-HLA interaction analysis showed strong binding of these epitopes with several common HLA molecules. The in silico immune simulation and population coverage analysis of the vaccine showed its potential to evoke strong immune responses.. Further, the interaction between vaccine and immune was evaluated by docking and simulation, revealing high affinity and complex stability. Codon adaptation and in silico cloning revealed higher expression of vaccine in E. coli K12 expression system. CONCLUSIONS Conclusively, the findings of the present study suggest that the designed novel multi-epitopic vaccine holds potential for further experimental validation against the pathogen.
Collapse
Affiliation(s)
- Jyotirmayee Dey
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, India
| | - Soumya Ranjan Mahapatra
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, India
| | - S Lata
- Kalinga Institute of Dental Sciences, KIIT Deemed to Be University, Bhubaneswar, India
| | - Shubhransu Patro
- Kalinga Institute of Medical Sciences, KIIT Deemed to Be University, Bhubaneswar, India
| | - Namrata Misra
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, India.,KIIT-Technology Business Incubator (KIIT-TBI), Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, India
| | - Mrutyunjay Suar
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, India.,KIIT-Technology Business Incubator (KIIT-TBI), Kalinga Institute of Industrial Technology (KIIT), Deemed to Be University, Bhubaneswar, India
| |
Collapse
|
47
|
Jann J, Drevelle O, Chen XG, Auclair-Gilbert M, Soucy G, Faucheux N, Fortier LC. Rapid antibacterial activity of anodized aluminum-based materials impregnated with quaternary ammonium compounds for high-touch surfaces to limit transmission of pathogenic bacteria. RSC Adv 2021; 11:38172-38188. [PMID: 35498065 PMCID: PMC9044312 DOI: 10.1039/d1ra07159a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/22/2021] [Indexed: 01/01/2023] Open
Abstract
Infections caused by multidrug-resistant bacteria are a major public health problem. Their transmission is strongly linked to cross contamination via inert surfaces, which can serve as reservoirs for pathogenic microorganisms. To address this problem, antibacterial materials applied to high-touch surfaces have been developed. However, reaching a rapid and lasting effectiveness under real life conditions of use remains challenging. In the present paper, hard-anodized aluminum (AA) materials impregnated with antibacterial agents (quaternary ammonium compounds (QACs) and/or nitrate silver (AgNO3)) were prepared and characterized. The thickness of the anodized layer was about 50 μm with pore diameter of 70 nm. AA with QACs and/or AgNO3 had a water contact angle varying between 45 and 70°. The antibacterial activity of the materials was determined under different experimental settings to better mimic their use, and included liquid, humid, and dry conditions. AA-QAC surfaces demonstrated excellent efficiency, killing >99.9% of bacteria in 5 min on a wide range of Gram-positive (Staphylococcus aureus, Clostridioides difficile, vancomycin-resistant Enterococcus faecium) and Gram-negative (streptomycin-resistant Salmonella typhimurium and encapsulated Klebsiella pneumoniae) pathogens. AA-QACs showed a faster antibacterial activity (from 0.25 to 5 min) compared with antibacterial copper used as a reference (from 15 min to more than 1 h). We show that to maintain their high performance, AA-QACs should be used in low humidity environments and should be cleaned with solutions composed of QACs. Altogether, AA-QAC materials constitute promising candidates to prevent the transmission of pathogenic bacteria on high-touch surfaces.
Collapse
Affiliation(s)
- Jessica Jann
- Department of Chemical and Biotechnological Engineering, Faculty of Engineering, Université de Sherbrooke 2500 boul. de l'Université Sherbrooke Québec J1K 2R1 Canada .,Clinical Research Center of Centre Hospitalier Universitaire de Sherbrooke 12e Avenue N Sherbrooke Québec J1H 5N4 Canada.,Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke 3201 rue Jean Mignault Sherbrooke Québec J1E 4K8 Canada
| | - Olivier Drevelle
- Department of Chemical and Biotechnological Engineering, Faculty of Engineering, Université de Sherbrooke 2500 boul. de l'Université Sherbrooke Québec J1K 2R1 Canada
| | - X Grant Chen
- Department of Applied Science, University of Quebec in Chicoutimi Saguenay Quebec G7H 2B1 Canada
| | | | - Gervais Soucy
- Department of Chemical and Biotechnological Engineering, Faculty of Engineering, Université de Sherbrooke 2500 boul. de l'Université Sherbrooke Québec J1K 2R1 Canada
| | - Nathalie Faucheux
- Department of Chemical and Biotechnological Engineering, Faculty of Engineering, Université de Sherbrooke 2500 boul. de l'Université Sherbrooke Québec J1K 2R1 Canada .,Clinical Research Center of Centre Hospitalier Universitaire de Sherbrooke 12e Avenue N Sherbrooke Québec J1H 5N4 Canada
| | - Louis-Charles Fortier
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke 3201 rue Jean Mignault Sherbrooke Québec J1E 4K8 Canada
| |
Collapse
|
48
|
Assoni L, Girardello R, Converso TR, Darrieux M. Current Stage in the Development of Klebsiella pneumoniae Vaccines. Infect Dis Ther 2021; 10:2157-2175. [PMID: 34476772 PMCID: PMC8412853 DOI: 10.1007/s40121-021-00533-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/24/2021] [Indexed: 01/14/2023] Open
Abstract
Klebsiella pneumoniae is a bacterium capable of colonizing mucous membranes, causing serious infections. Widespread antibiotic resistance in K. pneumoniae—either through intrinsic mechanisms or via acquisition from different species, especially in hospital environments—limits the therapeutic options against this pathogen, further aggravating the disease burden. To date, there are no vaccines available against K. pneumoniae infection. Although formulations based on capsular polysaccharides have been proposed, the high variability in capsular serotypes limits vaccine coverage. Recombinant vaccines based on surface exposed bacterial antigens are a promising alternative owing to their conservation among different serotypes and accessibility to the immune system. Many vaccine candidates have been proposed, some of which have reached clinical trials. The present review summarizes the current status of K. pneumoniae vaccine development. Different strategies including whole cell vaccines, outer membrane vesicles (OMVs), ribosome, polysaccharide, lipopolysaccharide (LPS), and protein-based formulations are discussed. The contribution of antibody and cell-mediated responses is also presented. In summary, K. pneumoniae vaccines are feasible and a promising strategy to prevent infections and to reduce the antimicrobial resistance burden worldwide.
Collapse
Affiliation(s)
- Lucas Assoni
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Raquel Girardello
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Thiago Rojas Converso
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil
| | - Michelle Darrieux
- Laboratório de Biologia Molecular de Microrganismos, Universidade São Francisco, Bragança Paulista, Brazil.
| |
Collapse
|
49
|
Dunstan RA, Bamert RS, Belousoff MJ, Short FL, Barlow CK, Pickard DJ, Wilksch JJ, Schittenhelm RB, Strugnell RA, Dougan G, Lithgow T. Mechanistic Insights into the Capsule-Targeting Depolymerase from a Klebsiella pneumoniae Bacteriophage. Microbiol Spectr 2021; 9:e0102321. [PMID: 34431721 PMCID: PMC8552709 DOI: 10.1128/spectrum.01023-21] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 02/07/2023] Open
Abstract
The production of capsular polysaccharides by Klebsiella pneumoniae protects the bacterial cell from harmful environmental factors such as antimicrobial compounds and infection by bacteriophages (phages). To bypass this protective barrier, some phages encode polysaccharide-degrading enzymes referred to as depolymerases to provide access to cell surface receptors. Here, we characterized the phage RAD2, which infects K. pneumoniae strains that produce the widespread, hypervirulence-associated K2-type capsular polysaccharide. Using transposon-directed insertion sequencing, we have shown that the production of capsule is an absolute requirement for efficient RAD2 infection by serving as a first-stage receptor. We have identified the depolymerase responsible for recognition and degradation of the capsule, determined that the depolymerase forms globular appendages on the phage virion tail tip, and present the cryo-electron microscopy structure of the RAD2 capsule depolymerase at 2.7-Å resolution. A putative active site for the enzyme was identified, comprising clustered negatively charged residues that could facilitate the hydrolysis of target polysaccharides. Enzymatic assays coupled with mass spectrometric analyses of digested oligosaccharide products provided further mechanistic insight into the hydrolase activity of the enzyme, which, when incubated with K. pneumoniae, removes the capsule and sensitizes the cells to serum-induced killing. Overall, these findings expand our understanding of how phages target the Klebsiella capsule for infection, providing a framework for the use of depolymerases as antivirulence agents against this medically important pathogen. IMPORTANCE Klebsiella pneumoniae is a medically important pathogen that produces a thick protective capsule that is essential for pathogenicity. Phages are natural predators of bacteria, and many encode diverse "capsule depolymerases" which specifically degrade the capsule of their hosts, an exploitable trait for potential therapies. We have determined the first structure of a depolymerase that targets the clinically relevant K2 capsule and have identified its putative active site, providing hints to its mechanism of action. We also show that Klebsiella cells treated with a recombinant form of the depolymerase are stripped of capsule, inhibiting their ability to grow in the presence of serum, demonstrating the anti-infective potential of these robust and readily producible enzymes against encapsulated bacterial pathogens such as K. pneumoniae.
Collapse
Affiliation(s)
- Rhys A. Dunstan
- Infection and Immunity Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Rebecca S. Bamert
- Infection and Immunity Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Matthew J. Belousoff
- Infection and Immunity Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Francesca L. Short
- Infection and Immunity Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Christopher K. Barlow
- Monash Proteomics & Metabolomics Facility, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
| | - Derek J. Pickard
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Jonathan J. Wilksch
- Infection and Immunity Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Ralf B. Schittenhelm
- Monash Proteomics & Metabolomics Facility, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
| | - Richard A. Strugnell
- Department of Microbiology and Immunology, The Peter Doherty Institute, The University of Melbourne, Parkville, Australia
| | - Gordon Dougan
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Trevor Lithgow
- Infection and Immunity Program, Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| |
Collapse
|
50
|
Ali S, Alam M, Hasan GM, Hassan MI. Potential therapeutic targets of Klebsiella pneumoniae: a multi-omics review perspective. Brief Funct Genomics 2021; 21:63-77. [PMID: 34448478 DOI: 10.1093/bfgp/elab038] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 11/15/2022] Open
Abstract
The multidrug resistance developed in many organisms due to the prolonged use of antibiotics has been an increasing global health crisis. Klebsiella pneumoniae is a causal organism for various infections, including respiratory, urinary tract and biliary diseases. Initially, immunocompromised individuals are primarily affected by K. pneumoniae. Due to the emergence of hypervirulent strains recently, both healthy and immunocompetent individuals are equally susceptible to K. pneumoniae infections. The infections caused by multidrug-resistant and hypervirulent K. pneumoniae strains are complicated to treat, illustrating an urgent need to develop novel and more practical approaches to combat the pathogen. We focused on the previously performed high-throughput analyses by other groups to discover several novel enzymes that may be considered attractive drug targets of K. pneumoniae. These targets qualify most of the selection criteria for drug targeting, including an absence of its homolog's gene in the host. The capsule, lipopolysaccharide, fimbriae, siderophores and essential virulence factors facilitate the pathogen entry, infection and survival inside the host. This review discusses K. pneumoniae pathophysiology, including its virulence determinants and further the potential drug targets that might facilitate the discovery of novel drugs and effective treatment regimens shortly.
Collapse
Affiliation(s)
- Sabeeha Ali
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar New Delhi 110025, India
| | - Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar New Delhi 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar New Delhi 110025, India
| |
Collapse
|