1
|
Lavergne JP, Page A, Polard P, Campo N, Grangeasse C. Quantitative phosphoproteomic reveals that the induction of competence modulates protein phosphorylation in Streptococcus pneumonaie. J Proteomics 2025; 315:105399. [PMID: 39921128 DOI: 10.1016/j.jprot.2025.105399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/19/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Competence in the pathogenic bacterium Streptococcus pneumoniae (S. pneumoniae) is a developmental genetic program that is key for natural genetic transformation and consequently bacterial horizontal gene transfer. Phosphoproteomic studies have revealed that protein phosphorylation on serine, threonine and tyrosine residues is a widespread regulatory post-translational modification in bacteria. In this study, we performed quantitative proteomic and phosphoproteomic analyses on S. pneumoniae as a function of competence induction. To calculate peptide abundance ratios between non-competent and competent samples we used dimethyl-tag labeling. Titanium dioxide (TiO2) beads were used for phosphopeptide enrichment and samples were analysed by LC-MS/MS. Our proteome analysis covers approximatively 63 % of the total bacterial protein content, identifying 82 proteins with significantly different abundance ratios, including some not previously linked to competence. 248 phosphopeptides were identified including 47 having different abundance ratios. Notably, the proteins with a change in phosphorylation in competent cells are different from the proteins with a change in expression, highlighting different pathways induced by competence and regulated by phosphorylation. This is the first report that phosphorylation of some proteins is regulated during competence in Streptococcus pneumoniae, a key pathway for the bacteria to evade vaccines or acquire antibiotic resistance. SIGNIFICANCE: S. pneumoniae is a prominent model for the study of competence that governs the development of natural genetic transformation. The latter largely accounts for the spread of antibiotic resistance and vaccine evasion in pneumococcal isolates. Many proteins specifically expressed during competence have been identified and extensively studied. However, the potential contribution of post-translational modifications, and notably phosphorylation, during the development of competence has never been investigated. In this study, we used a quantitative phosphoproteomic approach to determine both the protein expression and the protein phosphorylation patterns. Comparison of these patterns allows to highlight a series of proteins that are differentially phosphorylated in the two conditions. This result opens new avenues to decipher new regulatory pathways induced by competence and that are potentially key for natural genetic transformation. Interfering with theses regulatory pathways could represent a promising strategy to combat antibiotic resistance in the future.
Collapse
Affiliation(s)
- Jean-Pierre Lavergne
- Molecular Microbiology and Structural Biochemistry (MMSB), CNRS UMR 5086, Univiversité Lyon 1, Lyon 69007, France
| | - Adeline Page
- Protein Science Platform, SFR BioSciences, CNRS UAR3444, INSERM US8, Univiversité Lyon 1, ENS de Lyon, 69007 Lyon, France
| | - Patrice Polard
- Laboratoire de Microbiologie et Génétique Moléculaires, UMR5100, Centre de Biologie Intégrative, Centre Nationale de la Recherche Scientifique, 31062 Toulouse, France; Université Paul Sabatier (Toulouse III), 31062 Toulouse, France
| | - Nathalie Campo
- Laboratoire de Microbiologie et Génétique Moléculaires, UMR5100, Centre de Biologie Intégrative, Centre Nationale de la Recherche Scientifique, 31062 Toulouse, France; Université Paul Sabatier (Toulouse III), 31062 Toulouse, France.
| | - Christophe Grangeasse
- Molecular Microbiology and Structural Biochemistry (MMSB), CNRS UMR 5086, Univiversité Lyon 1, Lyon 69007, France.
| |
Collapse
|
2
|
Shahid S, Balka M, Lundin D, Daley DO, Sjöberg B, Rozman Grinberg I. NrdR in Streptococcus and Listeria spp.: DNA Helix Phase Dependence of the Bacterial Ribonucleotide Reductase Repressor. Mol Microbiol 2025; 123:406-419. [PMID: 39967291 PMCID: PMC12051234 DOI: 10.1111/mmi.15349] [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: 08/22/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/20/2025]
Abstract
NrdR is a universal transcriptional repressor of bacterial genes coding for ribonucleotide reductases (RNRs), essential enzymes that provide DNA building blocks in all living cells. Despite its bacterial prevalence, the NrdR mechanism has been scarcely studied. We report the biochemical, biophysical, and bioinformatical characterization of NrdR and its binding sites from two major bacterial pathogens of the phylum Bacillota Listeria monocytogenes and Streptococcus pneumoniae. NrdR consists of a Zn-ribbon domain followed by an ATP-cone domain. We show that it forms tetramers that bind to DNA when loaded with ATP and dATP, but if loaded with only ATP, NrdR forms various oligomeric complexes unable to bind DNA. The DNA-binding site in L. monocytogenes is a pair of NrdR boxes separated by 15-16 bp, whereas in S. pneumoniae, the NrdR boxes are separated by unusually long spacers of 25-26 bp. This observation triggered a comprehensive binding study of four NrdRs from L. monocytogenes, S. pneumoniae, Escherichia coli, and Streptomyces coelicolor to a series of dsDNA fragments where the NrdR boxes were separated by 12-27 bp. The in vitro results were confirmed in vivo in E. coli and revealed that NrdR binds most efficiently when there is an integer number of DNA turns between the center of the two NrdR boxes. The study facilitates the prediction of NrdR binding sites in bacterial genomes and suggests that the NrdR mechanism is conserved throughout the bacterial domain. It sheds light on RNR regulation in Listeria and Streptococcus, and since NrdR does not occur in eukaryotes, opens a way to the development of novel antibiotics.
Collapse
Affiliation(s)
- Saher Shahid
- Department of Biochemistry and BiophysicsStockholm UniversityStockholmSweden
| | - Mateusz Balka
- Department of Biochemistry and BiophysicsStockholm UniversityStockholmSweden
| | - Daniel Lundin
- Department of Biochemistry and BiophysicsStockholm UniversityStockholmSweden
| | - Daniel O. Daley
- Department of Biochemistry and BiophysicsStockholm UniversityStockholmSweden
| | - Britt‐Marie Sjöberg
- Department of Biochemistry and BiophysicsStockholm UniversityStockholmSweden
| | | |
Collapse
|
3
|
Lv J, Wan H, Yu D, Zhou H, Wang W, Wan H. Alleviating penicillin-resistant Streptococcus pneumoniae‑induced lung epithelial cell injury: mechanistic insights into effects of the optimized combination of main components from Yinhuapinggan granules. BMC Infect Dis 2025; 25:565. [PMID: 40254610 PMCID: PMC12010621 DOI: 10.1186/s12879-025-10951-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025] Open
Abstract
OBJECTIVE Penicillin-resistant Streptococcus pneumoniae (PRSP), for which novel treatment medicines are required, has expanded extensively due to the overuse of antibiotics. This study aimed to detect the optimal ratio of the combination of the main components based on Yinhuapinggan granules (YHPG) to generate novel treatment concepts for PRSP-induced lung injury. METHODS Three representative main components: chlorogenic acid (C), amygdalin (A), and puerarin (P) were selected, and the optimal combination of these three components was determined by an orthogonal experiment. Investigations were conducted on the potential mechanisms underlying the protective effect of this optimized combination against PRSP-induced lung epithelial cell damage. Meanwhile, the bacteriostatic effect was further explored through the optimized combination of these natural products combined with penicillin G (PG). RESULTS The optimized combination CAP (C: 16 µg/mL, A: 24 µg/mL, P: 24 µg/mL) screened by the orthogonal experimental design reduced cell damage in a model of human lung epithelial cells infected by PRSP, and the combination of CAP and PG had a synergistic effect. At the cellular level, CAP attenuated lung epithelial cell injury by modulating the TLRs/MyD88 inflammatory pathway. At the bacterial level, CAP modulated the virulence and drug resistance of PRSP, resulting in enhanced bacterial inhibition by the combination of CAP and PG. CONCLUSION Taken together, our results suggest that CAP can modulate or synergize with PG to modulate the TLRs/MyD88 pathway and attenuate PRSP-induced lung injury, and can be used as a potential drug for treating PRSP infection.
Collapse
Affiliation(s)
- Jiangbo Lv
- College of Chinese Medicine for Cardiovascular-Cranial Disease, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Chinese Medicine for Cardiovascular and Cerebrovascular Disease, Hangzhou, China
| | - Haofang Wan
- College of Chinese Medicine for Cardiovascular-Cranial Disease, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Chinese Medicine for Cardiovascular and Cerebrovascular Disease, Hangzhou, China
| | - Daojun Yu
- Hangzhou First People's Hospital, Hangzhou, China.
| | - Huifen Zhou
- College of Chinese Medicine for Cardiovascular-Cranial Disease, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Chinese Medicine for Cardiovascular and Cerebrovascular Disease, Hangzhou, China
| | - Wenba Wang
- College of Chinese Medicine for Cardiovascular-Cranial Disease, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Chinese Medicine for Cardiovascular and Cerebrovascular Disease, Hangzhou, China
| | - Haitong Wan
- College of Chinese Medicine for Cardiovascular-Cranial Disease, Zhejiang Chinese Medical University, Hangzhou, China.
- Zhejiang Key Laboratory of Chinese Medicine for Cardiovascular and Cerebrovascular Disease, Hangzhou, China.
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China.
| |
Collapse
|
4
|
García E. Structure, Function, and Regulation of LytA: The N-Acetylmuramoyl-l-alanine Amidase Driving the "Suicidal Tendencies" of Streptococcus pneumoniae-A Review. Microorganisms 2025; 13:827. [PMID: 40284663 PMCID: PMC12029793 DOI: 10.3390/microorganisms13040827] [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: 03/10/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
Streptococcus pneumoniae (pneumococcus) is a significant human pathogen responsible for a range of diseases from mild infections to invasive pneumococcal diseases, particularly affecting children, the elderly, and immunocompromised individuals. Despite pneumococcal conjugate vaccines having reduced disease incidence, challenges persist due to serotype diversity, vaccine coverage gaps, and antibiotic resistance. This review highlights the role of LytA, a key autolysin (N-acetylmuramoyl-l-alanine amidase), in pneumococcal biology. LytA regulates autolysis, contributes to inflammation, and biofilm formation, and impairs bacterial clearance. It also modulates complement activation, aiding immune evasion. LytA expression is influenced by environmental signals and genetic regulation and is tied to competence for genetic transformation, which is an important virulence trait, particularly in meningitis. With the increase in antibiotic resistance, LytA has emerged as a potential therapeutic target. Current research explores its use in bacteriolytic therapies, vaccine development, and synergistic antibiotic strategies. Various compounds, including synthetic peptides, plant extracts, and small molecules, have been investigated for their ability to trigger LytA-mediated bacterial lysis. Future directions include the development of novel anti-pneumococcal interventions leveraging LytA's properties while overcoming vaccine efficacy and resistance-related challenges. Human challenge models and animal studies continue to deepen our understanding of pneumococcal pathogenesis and potential treatment strategies.
Collapse
Affiliation(s)
- Ernesto García
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, 28040 Madrid, Spain
| |
Collapse
|
5
|
Olivella-Gomez J, Lozada J, Serrano-Mayorga CC, Méndez-Castillo L, Acosta-González A, Viñán Garcés AE, Bustos IG, Ibáñez-Prada ED, Fuentes YV, Crispin AM, Garcia-Garcia EY, Santana E, Josa DF, Pulido Saenz J, Rodíguez-Castaño GP, Rodríguez Orjuela JA, Jaimes D, Tettelin H, Orihuela CJ, Reyes LF. The relation of nasopharyngeal colonization by Streptococcus pneumoniae in comorbid adults with unfavorable outcomes in a low-middle income country. PLoS One 2025; 20:e0318320. [PMID: 39937814 PMCID: PMC11819510 DOI: 10.1371/journal.pone.0318320] [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: 10/11/2024] [Accepted: 01/14/2025] [Indexed: 02/14/2025] Open
Abstract
PURPOSE Streptococcus pneumoniae (Spn) is the primary bacterial cause of lower respiratory tract infections (LRTI) globally, particularly impacting older adults and children. While Spn colonization in children is linked to LRTI, its prevalence, and consequences in adults with comorbidities remain uncertain. This study aims to provide novel data in that regard. METHODS This prospective study of outpatient adults with chronic diseases was conducted in Colombia. Data on demographics, vaccination, and clinical history was collected in a RedCap database. Nasopharyngeal aspirate samples were examined for Spn colonization using traditional cultures and quantitative-real time polymerase chain reaction (q-rtPCR). Patients were followed for 18 months, with colonization prevalence calculated and factors influencing colonization and its impact on clinical outcomes analyzed through logistic regressions. RESULTS 810 patients were enrolled, with 10.1% (82/810) identified as colonized. The mean (SD) age was 62 years (±15), and 48.6% (394/810) were female. Major comorbidities included hypertension (52.2% [423/810]), cardiac conditions (31.1% [252/810]), and chronic kidney disease (17.4% [141/810]). Among all, 31.6% (256/810) received the influenza vaccine in the previous year, and 10.7% (87/810) received anti-Spn vaccines. Chronic kidney disease (OR 95% CI; 2.48 [1.01-6.15], p = 0.04) and chronic cardiac diseases (OR 95% CI; 1.62 [0.99-2.66], p = 0.05) were independently associated with Spn colonization. However, colonization was not associated with the development of LRTI (OR 95%CI; 0.64 [0.14-2.79], p = 0.55) or unfavorable outcomes (OR 95% CI;1.17 [0.14-2.79], p = 0.54) during follow-up. CONCLUSIONS Chronic kidney and cardiac diseases are independently associated with Spn colonization. However, Spn colonization was not associated with LRTI/unfavorable outcomes in adult patients with chronic comorbidities in our cohort.
Collapse
Affiliation(s)
- Juan Olivella-Gomez
- School of Medicine, Universidad de La Sabana, Chía, Colombia
- Unisabana Center for Translational Science, Universidad de La Sabana, Chía, Colombia
| | - Julián Lozada
- School of Medicine, Universidad de La Sabana, Chía, Colombia
- Clínica Universidad de La Sabana, Chía, Colombia
- Biosciences PhD, Engineering Faculty, Universidad de La Sabana, Chía, Colombia
| | - Cristian C. Serrano-Mayorga
- School of Medicine, Universidad de La Sabana, Chía, Colombia
- Unisabana Center for Translational Science, Universidad de La Sabana, Chía, Colombia
- Clínica Universidad de La Sabana, Chía, Colombia
- Biosciences PhD, Engineering Faculty, Universidad de La Sabana, Chía, Colombia
| | | | - Alejandro Acosta-González
- Unisabana Center for Translational Science, Universidad de La Sabana, Chía, Colombia
- Grupo de Investigación en Bioprospección (G.I.B.P.), Faculty of Engineering, Universidad de La Sabana, Chía, Colombia
| | - André Emilio Viñán Garcés
- Unisabana Center for Translational Science, Universidad de La Sabana, Chía, Colombia
- Clínica Universidad de La Sabana, Chía, Colombia
| | - Ingrid G. Bustos
- School of Medicine, Universidad de La Sabana, Chía, Colombia
- Unisabana Center for Translational Science, Universidad de La Sabana, Chía, Colombia
- Biosciences PhD, Engineering Faculty, Universidad de La Sabana, Chía, Colombia
| | - Elsa D. Ibáñez-Prada
- Unisabana Center for Translational Science, Universidad de La Sabana, Chía, Colombia
- Clínica Universidad de La Sabana, Chía, Colombia
| | - Yuli V. Fuentes
- School of Medicine, Universidad de La Sabana, Chía, Colombia
| | - Ana M. Crispin
- Unisabana Center for Translational Science, Universidad de La Sabana, Chía, Colombia
| | | | | | - Diego F. Josa
- School of Medicine, Universidad de La Sabana, Chía, Colombia
- Fundación Clínica Shaio, Department of Clinical Laboratory and Pathology, Molecular Biology - Microbiology Area Bogotá, Bogotá, Colombia
| | | | - Gina Paola Rodíguez-Castaño
- Grupo de Investigación en Bioprospección (G.I.B.P.), Faculty of Engineering, Universidad de La Sabana, Chía, Colombia
| | | | - Diego Jaimes
- School of Medicine, Universidad de La Sabana, Chía, Colombia
- Unisabana Center for Translational Science, Universidad de La Sabana, Chía, Colombia
| | - Hervé Tettelin
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Carlos J. Orihuela
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Luis Felipe Reyes
- School of Medicine, Universidad de La Sabana, Chía, Colombia
- Unisabana Center for Translational Science, Universidad de La Sabana, Chía, Colombia
- Clínica Universidad de La Sabana, Chía, Colombia
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
6
|
Rafei R, Zaylaa M, Diab M, Kassem II, El Omari K, Halimeh FB, El Moujaber G, Achour A, Ismail B, Mallat H, Hamze M, Dabboussi F, Osman M. Nasopharyngeal Carriage, Antimicrobial Resistance, and Serotype Distribution of Streptococcus pneumoniae in Children Under Five in Lebanon: Baseline Data Prior to PCV13 Introduction. Antibiotics (Basel) 2025; 14:168. [PMID: 40001412 PMCID: PMC11851980 DOI: 10.3390/antibiotics14020168] [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: 12/30/2024] [Revised: 01/20/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND The nasopharyngeal carriage of Streptococcus pneumoniae can be the source of transmission between humans and the starting step towards invasive pneumococcal diseases. Data on the carriage of pneumococci in children before and after the pneumococcal conjugate vaccines (PCV) integration in a country are essential for monitoring any change in pneumococcal carriage serotypes and their antimicrobial-resistance profiles. METHODS We investigated the epidemiology of S. pneumoniae carriage among children younger than five years old in Tripoli, Lebanon, in 2016, the same year of integration of PCV13 in the country's Expanded Program on Immunization. RESULTS Of 104 participating children, 57 (54.8%) gave a positive culture for S. pneumoniae. Antimicrobial susceptibility testing revealed that 26.3% of isolates were multidrug-resistant. Resistance was detected mainly against oxacillin (77.2%), tetracycline (29.8%), erythromycin (22.8%), trimethoprim-sulfamethoxazole (22.8%), clindamycin (19.3%), minocycline (19.3%), and teicoplanin (1.8%). Serotyping analysis identified 14 distinct serotypes, with only 31.3% and 50% of isolates corresponding to vaccine serotypes covered by PCV13 and PCV20, respectively. The most common serotypes were 11A, 19F, 23A, and those of serogroup 24 (Sg24) accounted for 37.5% of the serotyped isolates. CONCLUSIONS Our findings have revealed the circulation of a pool of pneumococci isolates with high levels of antibiotic resistance and different degrees of likelihood of causing invasive diseases in children under five years old in Tripoli in 2016. The overall limited PCV13 vaccine coverage in this study highlighted the need for vaccines with greater coverage in the immunization programs in Lebanon. Longitudinal national studies investigating the carriage of pneumococci in children are required to further assess the impact of the PCV vaccine on pneumococci carriage in children and steer new vaccine development.
Collapse
Affiliation(s)
- Rayane Rafei
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School for Science and Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| | - Mazen Zaylaa
- Faculty of Medicine, Beirut Arab University, Beirut, Lebanon
| | - Mohamad Diab
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School for Science and Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| | - Issmat I. Kassem
- Center for Food Safety, Department of Food Science and Technology, University of Georgia, Griffin, GA 30223-1797, USA
- Faculty of Agricultural and Food Sciences, American University of Beirut, Riad El Solh, Beirut, Lebanon
| | - Khaled El Omari
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School for Science and Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
- Quality Control Center Laboratories at the Chamber of Commerce, Industry and Agriculture of Tripoli and North Lebanon, Tripoli, Lebanon
| | - Fatima B. Halimeh
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School for Science and Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| | - Grace El Moujaber
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School for Science and Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| | - Afaf Achour
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School for Science and Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| | - Bassel Ismail
- College of Health and Medical Technologies, Alayen Iraqi University (AUIQ), Thi Qar, Iraq
| | - Hassan Mallat
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School for Science and Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| | - Monzer Hamze
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School for Science and Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| | - Fouad Dabboussi
- Laboratoire Microbiologie Santé et Environnement (LMSE), Doctoral School for Science and Technology, Faculty of Public Health, Lebanese University, Tripoli, Lebanon
| | - Marwan Osman
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
7
|
Scanlan PD, Baquero F, Levin BR. Short-sighted evolution of virulence for invasive gut microbes: From hypothesis to tests. Proc Natl Acad Sci U S A 2024; 121:e2409905121. [PMID: 39570365 PMCID: PMC11626195 DOI: 10.1073/pnas.2409905121] [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: 11/22/2024] Open
Abstract
Why microbes harm their hosts is a fundamental question in evolutionary biology with broad relevance to our understanding of infectious diseases. Several hypotheses have been proposed to explain this "evolution of virulence." In this perspective, we reexamine one of these hypotheses in the specific context of the human gut microbiome, namely short-sighted evolution. According to the short-sighted evolution hypothesis, virulence is a product of niche expansion within a colonized host, whereby variants of commensal microbes establish populations in tissues and sites where the infection causes morbidity or mortality. This evolution is short-sighted in that the evolved variants that infect those tissues and sites are not transmitted to other hosts. The specific hypothesis that we propose is that some bacteria responsible for invasive infections and disease are the products of the short-sighted evolution of commensal bacteria residing in the gut microbiota. We present observations in support of this hypothesis and discuss the challenges inherent in assessing its general application to infections and diseases associated with specific members of the gut microbiota. We then describe how this hypothesis can be tested using genomic data and animal model experiments and outline how such studies will serve to provide fundamental information about both the evolution and genetic basis of virulence, and the bacteria of intensively studied yet poorly understood habitats including the gut microbiomes of humans and other mammals.
Collapse
Affiliation(s)
- Pauline D. Scanlan
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
- School of Microbiology, University College Cork, CorkT12 Y337, Ireland
| | - Fernando Baquero
- Servicio de Microbiología, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid28034, Spain
- Centro de Investigación Médica en Red, Epidemiología y Salud Pública, Madrid28007, Spain
| | - Bruce R. Levin
- Department of Biology, Emory University, Atlanta, GA30322
| |
Collapse
|
8
|
Waz NT, Milani B, Assoni L, Coelho GR, Sciani JM, Parisotto T, Ferraz LFC, Hakansson AP, Converso TR, Darrieux M. Pneumococcal surface protein A (PspA) prevents killing of Streptococcus pneumoniae by indolicidin. Sci Rep 2024; 14:23517. [PMID: 39384882 PMCID: PMC11464550 DOI: 10.1038/s41598-024-73564-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 09/18/2024] [Indexed: 10/11/2024] Open
Abstract
Pneumococcal surface protein A (PspA) is an important virulence factor in Streptococcus pneumoniae that binds to lactoferrin and protects the bacterium from the bactericidal action of lactoferricins-cationic peptides released upon lactoferrin proteolysis. The present study investigated if PspA can prevent killing by another cationic peptide, indolicidin. PspA-negative pneumococci were more sensitive to indolicidin-induced killing than bacteria expressing PspA, suggesting that PspA prevents the bactericidal action of indolicidin. Similarly, chemical removal of choline-binding proteins increased sensitivity to indolicidin. The absence of capsule and PspA had an additive effect on pneumococcal killing by the AMP. Furthermore, anti-PspA antibodies enhanced the bactericidal effect of indolicidin on pneumococci, while addition of soluble PspA fragments competitively inhibited indolicidin action. Previous in silico analysis suggests a possible interaction between PspA and indolicidin. Thus, we hypothesize that PspA acts by sequestering indolicidin and preventing it from reaching the bacterial membrane. A specific interaction between PspA and indolicidin was demonstrated by mass spectrometry, confirming that PspA can actively bind to the AMP. These results reinforce the vaccine potential of PspA and suggest a possible mechanism of innate immune evasion employed by pneumococci, which involves binding to cationic peptides and hindering their ability to damage the bacterial membranes.
Collapse
Affiliation(s)
- Natalha T Waz
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | - Barbara Milani
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | - Lucas Assoni
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | | | - Juliana M Sciani
- Laboratório de Produtos Naturais, Universidade São Francisco, Bragança Paulista, Brazil
| | - Thaís Parisotto
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | - Lucio F C Ferraz
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | - Anders P Hakansson
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University, Lund, Sweden
| | - Thiago R Converso
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil
| | - Michelle Darrieux
- Laboratório de Microbiologia Molecular e Clínica, Universidade São Francisco, Bragança Paulista, Brazil.
| |
Collapse
|
9
|
Shen K, Miao W, Zhu L, Hu Q, Ren F, Dong X, Tong H. A 3'UTR-derived small RNA represses pneumolysin synthesis and facilitates pneumococcal brain invasion. Commun Biol 2024; 7:1130. [PMID: 39271946 PMCID: PMC11399405 DOI: 10.1038/s42003-024-06845-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Pneumolysin (Ply) of Streptococcus pneumoniae (pneumococcus) at relatively high and low levels facilitates pneumococcal invasion into the lung and brain, respectively; however, the regulatory mechanisms of Ply expression are poorly understood. Here, we find that a small RNA plyT, processed from the 3'UTR of the ply operon, is expressed higher in anaerobically- than in statically-cultured pneumococcus D39. Using bioinformatic, biochemical and genetic approaches, we reveal that PlyT inhibits Ply synthesis and hemolytic activities by pairing with an RBS-embedded intergenic region of the ply operon. The RNA-binding protein SPD_1558 facilitates the pairing. Importantly, PlyT inhibition of Ply synthesis is stronger in anaerobic culture and leads to lower Ply abundance. Deletion of plyT decreases the number of pneumococci in the infected mouse brain and reduces the virulence, demonstrating that PlyT-regulated lower Ply in oxygen-void microenvironments, such as the blood, is important for pneumococcus to cross the blood-brain barrier and invade the brain. PlyT-mediated repression of Ply synthesis at anoxic niches is also verified in pneumococcal serotype 4 and 14 strains; moreover, the ply operon with a 3'UTR-embedded plyT, and the pairing sequences of IGR and plyT are highly conserved among pneumococcal strains, implying PlyT-regulated Ply synthesis might be widely employed by pneumococcus.
Collapse
Affiliation(s)
- Kaiqiang Shen
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wenshuang Miao
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- School of Basic Medicine, Shenyang Medical College, Shenyang, China
| | - Lin Zhu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qingqing Hu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fu Ren
- School of Basic Medicine, Shenyang Medical College, Shenyang, China
| | - Xiuzhu Dong
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Huichun Tong
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
10
|
Panickar A, Manoharan A, Anbarasu A, Ramaiah S. Respiratory tract infections: an update on the complexity of bacterial diversity, therapeutic interventions and breakthroughs. Arch Microbiol 2024; 206:382. [PMID: 39153075 DOI: 10.1007/s00203-024-04107-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
Respiratory tract infections (RTIs) have a significant impact on global health, especially among children and the elderly. The key bacterial pathogens Streptococcus pneumoniae, Haemophilus influenzae, Klebsiella pneumoniae, Staphylococcus aureus and non-fermenting Gram Negative bacteria such as Acinetobacter baumannii and Pseudomonas aeruginosa are most commonly associated with RTIs. These bacterial pathogens have evolved a diverse array of resistance mechanisms through horizontal gene transfer, often mediated by mobile genetic elements and environmental acquisition. Treatment failures are primarily due to antimicrobial resistance and inadequate bacterial engagement, which necessitates the development of alternative treatment strategies. To overcome this, our review mainly focuses on different virulence mechanisms and their resulting pathogenicity, highlighting different therapeutic interventions to combat resistance. To prevent the antimicrobial resistance crisis, we also focused on leveraging the application of artificial intelligence and machine learning to manage RTIs. Integrative approaches combining mechanistic insights are crucial for addressing the global challenge of antimicrobial resistance in respiratory infections.
Collapse
Affiliation(s)
- Avani Panickar
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Anand Manoharan
- Infectious Diseases Medical and Scientific Affairs, GlaxoSmithKline (GSK), Worli, Maharashtra, India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
- Department of Bio-Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
11
|
Jain SS, Singh VK, Kante RK, Jana SK, Patil RH. Current trends in development and manufacturing of higher-valent pneumococcal polysaccharide conjugate vaccine and its challenges. Biologicals 2024; 87:101784. [PMID: 39053122 DOI: 10.1016/j.biologicals.2024.101784] [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: 04/21/2024] [Revised: 06/05/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024] Open
Abstract
Pneumococcal conjugate vaccines (PCVs) have been developed to protect against pneumococcal diseases caused by the more than 100 serotypes of the bacterium Streptococcus pneumoniae. PCVs primarily prevent pneumococcal infections such as sepsis, bacteraemia, meningitis, otitis media, pneumonia, septicaemia, and sinusitis among infants, adults, elderly, and immunocompromised individuals. The current available PCVs only cover a limited number of serotypes, and there is an immense need for developing higher-valent PCVs that can protect against non-vaccine serotypes to overcome challenges like serotype replacement and antibiotic resistance. The main challenges for developing higher valent PCVs are the complexity of the manufacturing process comprising polysaccharide fermentation, purification, modification or sizing of multiple polysaccharides and conjugation between polysaccharides and carrier proteins, the stability of the conjugates, and the immunogenicity of the vaccine. Different manufacturing processes have been explored to produce higher valent PCVs using different serotypes of S. pneumoniae and conjugation with different carrier proteins. The global coverage of higher valent PCVs are still low, mainly due to the high cost and limited supply of the vaccine. This review focuses on the existing and emerging manufacturing processes and challenges associated with higher-valent pneumococcal PCV development.
Collapse
Affiliation(s)
- Shital S Jain
- Savitribai Phule Pune University, Department of Biotechnology, Pune, Maharashtra, 411007, India; Serum Institute of India Pvt. Ltd., Hadapsar, Pune, Maharashtra, 411028, India.
| | - Vikas K Singh
- Serum Institute of India Pvt. Ltd., Hadapsar, Pune, Maharashtra, 411028, India.
| | - Rajesh Kumar Kante
- Serum Institute of India Pvt. Ltd., Hadapsar, Pune, Maharashtra, 411028, India.
| | - Swapan Kumar Jana
- Serum Institute of India Pvt. Ltd., Hadapsar, Pune, Maharashtra, 411028, India.
| | - Rajendra H Patil
- Savitribai Phule Pune University, Department of Biotechnology, Pune, Maharashtra, 411007, India.
| |
Collapse
|
12
|
Wang Y, Shi G, Wang X, Xie Z, Gou J, Huang L, Huang H, You W, Wang R, Yang Y, Wang F, Zhu T, Zhao D. Preliminary Evaluation of the Safety and Immunogenicity of a Novel Protein-Based Pneumococcal Vaccine in Healthy Adults Aged 18-49: A Phase Ia Randomized, Double Blind, Placebo-Controlled Clinical Study. Vaccines (Basel) 2024; 12:827. [PMID: 39203953 PMCID: PMC11358999 DOI: 10.3390/vaccines12080827] [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: 06/13/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 09/03/2024] Open
Abstract
Background: Protein-based pneumococcal vaccines (PBPVs) may offer expanded protection against Streptococcus pneumoniae and tackle the antimicrobial resistance crisis in pneumococcal infections. This study examined the safety and immunogenicity in healthy adults vaccinated with three doses of a protein-based pneumococcal vaccine containing pneumococcal surface protein A (PspA) (PRX1, P3296 and P5668) and in combination with a recombinant detoxified pneumolysin protein (PlyLD). Methods: This phase Ia randomized, double blind, placebo-controlled clinical study enrolled healthy adults aged 18-49 years. The participants were randomized into experimental (low-dose, medium-dose, high-dose) and placebo groups in a ratio of 3:1. Three doses of investigational vaccine were given to the participants with an interval of two months. Safety endpoints included the occurrence of total adverse reactions, solicited local and systemic adverse reactions, unsolicited adverse reactions, serious adverse events (SAEs), and several laboratory parameters. Immunogenicity endpoints included geometric mean titers (GMT) of anti-PspA (PRX1, P3296 and P5668) and anti-PlyLD antibodies level as determined by ELISA, seropositivity rates of PspA and PlyLD antibodies (>4-fold increase) and neutralization activity of anti-Ply antibody in serum. Results: A total of 118 participants completed the study of three doses. The candidate PBPV was safe and well-tolerated in all experimental groups. No vaccine-related SAEs were observed in this study. Most solicited adverse reactions were mild and transient. The most frequently reported solicited adverse reactions in the medium- and high-dose groups was pain at the injection site, while in the low-dose group it was elevated blood pressure. The immunogenicity data showed a sharp increase in the GMT level of anti-PspA-RX1, anti-PspA-3296, anti-PspA-5668, and anti-PlyLD antibodies in serum. The results also showed that the elicited antibodies were dosage-dependent. The high-dose group showed a higher immune response against PspA-RX1, PspA-3296, PspA-5668, and PlyLD antigens. However, repeat vaccination did not increase the level of anti-PspA antibodies but the level of anti-PlyLD antibody. High seropositivity rates were also observed for anti-PspA-RX1, anti-PspA-3296, anti-PspA-5668, and anti-PlyLD antibodies. In addition, a significant difference in the GMT levels of anti-Ply antibody between the high-, medium-, and low-dose groups post each vaccination were indicated by neutralization activity tests. Conclusions: The PBPV showed a safe and immunogenic profile in this clinical trial. Taking into consideration both safety and immunogenicity data, we propose a single dose of 50 µg (medium dose) of PBPV as the optimum approach in providing expanded protection against Streptococcus pneumoniae.
Collapse
Affiliation(s)
- Yanxia Wang
- Henan Center for Disease Control and Prevention, Zhengzhou 450016, China; (Y.W.); (Z.X.); (L.H.); (W.Y.)
| | - Gang Shi
- National Institutes for Food and Drug Control, Beijing 100050, China;
| | - Xue Wang
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (R.W.); (F.W.)
| | - Zhiqiang Xie
- Henan Center for Disease Control and Prevention, Zhengzhou 450016, China; (Y.W.); (Z.X.); (L.H.); (W.Y.)
| | - Jinbo Gou
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (R.W.); (F.W.)
| | - Lili Huang
- Henan Center for Disease Control and Prevention, Zhengzhou 450016, China; (Y.W.); (Z.X.); (L.H.); (W.Y.)
| | - Haitao Huang
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (R.W.); (F.W.)
| | - Wangyang You
- Henan Center for Disease Control and Prevention, Zhengzhou 450016, China; (Y.W.); (Z.X.); (L.H.); (W.Y.)
| | - Ruijie Wang
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (R.W.); (F.W.)
| | - Yongli Yang
- Department of Epidemiology and Health Statistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China;
| | - Feiyu Wang
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (R.W.); (F.W.)
| | - Tao Zhu
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (R.W.); (F.W.)
| | - Dongyang Zhao
- Henan Center for Disease Control and Prevention, Zhengzhou 450016, China; (Y.W.); (Z.X.); (L.H.); (W.Y.)
| |
Collapse
|
13
|
Cui L, Yang R, Huo D, Li L, Qu X, Wang J, Wang X, Liu H, Chen H, Wang X. Streptococcus pneumoniae extracellular vesicles aggravate alveolar epithelial barrier disruption via autophagic degradation of OCLN (occludin). Autophagy 2024; 20:1577-1596. [PMID: 38497494 PMCID: PMC11210924 DOI: 10.1080/15548627.2024.2330043] [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: 12/13/2023] [Revised: 02/25/2024] [Accepted: 03/09/2024] [Indexed: 03/19/2024] Open
Abstract
Streptococcus pneumoniae (S. pneumoniae) represents a major human bacterial pathogen leading to high morbidity and mortality in children and the elderly. Recent research emphasizes the role of extracellular vesicles (EVs) in bacterial pathogenicity. However, the contribution of S. pneumoniae EVs (pEVs) to host-microbe interactions has remained unclear. Here, we observed that S. pneumoniae infections in mice led to severe lung injuries and alveolar epithelial barrier (AEB) dysfunction. Infections of S. pneumoniae reduced the protein expression of tight junction protein OCLN (occludin) and activated macroautophagy/autophagy in lung tissues of mice and A549 cells. Mechanically, S. pneumoniae induced autophagosomal degradation of OCLN leading to AEB impairment in the A549 monolayer. S. pneumoniae released the pEVs that could be internalized by alveolar epithelial cells. Through proteomics, we profiled the cargo proteins inside pEVs and found that these pEVs contained many virulence factors, among which we identified a eukaryotic-like serine-threonine kinase protein StkP. The internalized StkP could induce the phosphorylation of BECN1 (beclin 1) at Ser93 and Ser96 sites, initiating autophagy and resulting in autophagy-dependent OCLN degradation and AEB dysfunction. Finally, the deletion of stkP in S. pneumoniae completely protected infected mice from death, significantly alleviated OCLN degradation in vivo, and largely abolished the AEB disruption caused by pEVs in vitro. Overall, our results suggested that pEVs played a crucial role in the spread of S. pneumoniae virulence factors. The cargo protein StkP in pEVs could communicate with host target proteins and even hijack the BECN1 autophagy initiation pathway, contributing to AEB disruption and bacterial pathogenicity.Abbreviations: AEB: alveolarepithelial barrier; AECs: alveolar epithelial cells; ATG16L1: autophagy related 16 like 1; ATP:adenosine 5'-triphosphate; BafA1: bafilomycin A1; BBB: blood-brain barrier; CFU: colony-forming unit; co-IP: co-immunoprecipitation; CQ:chloroquine; CTRL: control; DiO: 3,3'-dioctadecylox-acarbocyanineperchlorate; DOX: doxycycline; DTT: dithiothreitol; ECIS: electricalcell-substrate impedance sensing; eGFP: enhanced green fluorescentprotein; ermR: erythromycin-resistance expression cassette; Ery: erythromycin; eSTKs: eukaryotic-like serine-threoninekinases; EVs: extracellular vesicles; HA: hemagglutinin; H&E: hematoxylin and eosin; HsLC3B: human LC3B; hpi: hours post-infection; IP: immunoprecipitation; KD: knockdown; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LC/MS: liquid chromatography-mass spectrometry; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MVs: membranevesicles; NC:negative control; NETs:neutrophil extracellular traps; OD: optical density; OMVs: outer membrane vesicles; PBS: phosphate-buffered saline; pEVs: S.pneumoniaeextracellular vesicles; protK: proteinase K; Rapa: rapamycin; RNAi: RNA interference; S.aureus: Staphylococcusaureus; SNF:supernatant fluid; sgRNA: single guide RNA; S.pneumoniae: Streptococcuspneumoniae; S.suis: Streptococcussuis; TEER: trans-epithelium electrical resistance; moi: multiplicity ofinfection; TEM:transmission electron microscope; TJproteins: tight junction proteins; TJP1/ZO-1: tight junction protein1; TSA: tryptic soy agar; WB: western blot; WT: wild-type.
Collapse
Affiliation(s)
- Luqing Cui
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Ruicheng Yang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| | - Dong Huo
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Liang Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Xinyi Qu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Jundan Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Xinyi Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Hulin Liu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| | - Xiangru Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| |
Collapse
|
14
|
Baranchyk Y, Gestels Z, Van den Bossche D, Abdellati S, Britto Xavier B, Manoharan-Basil SS, Kenyon C. Effect of erythromycin residuals in food on the development of resistance in Streptococcus pneumoniae: an in vivo study in Galleria mellonella. PeerJ 2024; 12:e17463. [PMID: 38827315 PMCID: PMC11141549 DOI: 10.7717/peerj.17463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/05/2024] [Indexed: 06/04/2024] Open
Abstract
Background The use of antimicrobials to treat food animals may result in antimicrobial residues in foodstuffs of animal origin. The European Medicines Association (EMA) and World Health Organization (WHO) define safe antimicrobial concentrations in food based on acceptable daily intakes (ADIs). It is unknown if ADI doses of antimicrobials in food could influence the antimicrobial susceptibility of human-associated bacteria. Objectives This aim of this study was to evaluate if the consumption of ADI doses of erythromycin could select for erythromycin resistance in a Galleria mellonella model of Streptococcus pneumoniae infection. Methods A chronic model of S. pneumoniae infection in G. mellonella larvae was used for the experiment. Inoculation of larvae with S. pneumoniae was followed by injections of erythromycin ADI doses (0.0875 and 0.012 μg/ml according to EMA and WHO, respectively). Isolation of S. pneumoniae colonies was then performed on selective agar plates. Minimum inhibitory concentrations (MICs) of resistant colonies were measured, and whole genome sequencing (WGS) was performed followed by variant calling to determine the genetic modifications. Results Exposure to single doses of both EMA and WHO ADI doses of erythromycin resulted in the emergence of erythromycin resistance in S. pneumoniae. Emergent resistance to erythromycin was associated with a mutation in rplA, which codes for the L1 ribosomal protein and has been linked to macrolide resistance in previous studies. Conclusion In our in vivo model, even single doses of erythromycin that are classified as acceptable by the WHO and EMA induced significant increases in erythromycin MICs in S. pneumoniae. These results suggest the need to include the induction of antimicrobial resistance (AMR) as a significant criterion for determining ADIs.
Collapse
Affiliation(s)
- Yuliia Baranchyk
- UnivLyon, Université Claude Bernard Lyon 1, Lyon, France
- Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Zina Gestels
- Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | | | - Saïd Abdellati
- Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Basil Britto Xavier
- Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- Hospital Outbreak Support Team-HOST, Ziekenhuis Netwerk Antwerpen Middelheim, Antwerp, Belgium
| | | | - Chris Kenyon
- Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
15
|
Sari RF, Fadilah F, Maladan Y, Sarassari R, Safari D. A narrative review of genomic characteristics, serotype, immunogenicity, and vaccine development of Streptococcus pneumoniae capsular polysaccharide. Clin Exp Vaccine Res 2024; 13:91-104. [PMID: 38752009 PMCID: PMC11091432 DOI: 10.7774/cevr.2024.13.2.91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 03/31/2024] [Accepted: 04/05/2024] [Indexed: 05/18/2024] Open
Abstract
This narrative review describes genomic characteristic, serotyping, immunogenicity, and vaccine development of Streptococcus pneumoniae capsular polysaccharide (CPS). CPS is a primary virulence factor of S. pneumoniae. The genomic characteristics of S. pneumoniae CPS, including the role of biosynthetic gene and genetic variation within cps (capsule polysaccharide) locus which may lead to serotype replacement are still being investigated. One hundred unique serotypes of S. pneumoniae have been identified through various methods of serotyping using phenotypic and genotypic approach. The advantages and limitations of each method are various, emphasizing the need for accurate and comprehensive serotyping for effective disease surveillance and vaccine targeting. In addition, we elaborate the critical role of CPS in vaccine development by providing an overview of immunogenicity, ongoing research of pneumococcal vaccines, and the impact on disease burden.
Collapse
Affiliation(s)
- Ratna Fathma Sari
- Master’s Programme in Biomedical Sciences, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Indonesia
| | - Fadilah Fadilah
- Medical Chemistry Department, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Yustinus Maladan
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Indonesia
| | - Rosantia Sarassari
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Indonesia
| | - Dodi Safari
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Indonesia
| |
Collapse
|
16
|
Ríos E, Pérez M, Sanz JC, Delgado-Iribarren A, Rodríguez-Avial I. Efficacy of delafloxacin alone and in combination with cefotaxime against cefotaxime non-susceptible invasive isolates of Streptococcus pneumoniae. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2024; 37:158-162. [PMID: 38226580 PMCID: PMC10945109 DOI: 10.37201/req/107.2023] [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: 09/05/2023] [Revised: 11/20/2023] [Accepted: 12/19/2023] [Indexed: 01/17/2024]
Abstract
OBJECTIVE We assessed the in vitro activity of delafloxacin and the synergy between cefotaxime and delafloxacin among cefotaxime non-susceptible invasive isolates of Streptococcus pneumoniae (CNSSP). METHODS A total of 30 CNSSP (cefotaxime MIC > 0.5 mg/L) were studied. Serotyping was performed by the Pneumotest-Latex and Quellung reaction. Minimum inhibitory concentrations (MICs) of delafloxacin, levofloxacin, penicillin, cefotaxime, erythromycin and vancomycin were determined by gradient diffusion strips (GDS). Synergistic activity of delafloxacin plus cefotaxime against clinical S. pneumoniae isolates was evaluated by the GDS cross method. RESULTS Delafloxacin showed a higher pneumococcal activity than its comparator levofloxacin (MIC50, 0.004 versus 0.75 mg/L and MIC90, 0.047 versus >32 mg/L). Resistance to delafloxacin was identified in 7/30 (23.3%) isolates, belonging to serotypes 14 and 9V. Synergy between delafloxacin and cefotaxime was detected in 2 strains (serotypes 19A and 9V). Antagonism was not observed. Addition of delafloxacin increased the activity of cefotaxime in all isolates. Delafloxacin susceptibility was restored in 5/7 (71.4%) strains. CONCLUSIONS CNSSP showed a susceptibility to delafloxacin of 76.7%. Synergistic interactions between delafloxacin and cefotaxime were observed in vitro among CNSSP by GDS cross method.
Collapse
Affiliation(s)
- E Ríos
- Esther Ríos Dueñas, Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid. Plaza Ramón y Cajal, s/n 28040 Madrid, Spain.
| | | | | | | | | |
Collapse
|
17
|
de Felipe B, Aboza-García M, González-Galán V, Salamanca de la Cueva I, Martín-Quintero JA, Amil-Pérez B, Coronel-Rodríguez C, Palacios-Soria MÁ, García Ruiz-Santaquiteria MI, Torres-Sánchez MJ, Morón FJ, Cordero-Varela JA, Obando-Pacheco P, Obando I. Molecular epidemiology of pneumococcal carriage in children from Seville, following implementation of the PCV13 immunization program in Andalusia, Spain. ENFERMEDADES INFECCIOSAS Y MICROBIOLOGIA CLINICA (ENGLISH ED.) 2024; 42:172-178. [PMID: 37085445 DOI: 10.1016/j.eimce.2023.04.008] [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: 10/10/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 04/23/2023]
Abstract
INTRODUCTION The 13-valent pneumococcal conjugate vaccine (PCV13) universal vaccination programme was introduced in December 2016 in Andalusia. METHODS A cross-sectional study was conducted on the molecular epidemiology of pneumococcal nasopharyngeal colonization. A total of 397 healthy children were recruited from primary healthcare centres in Seville for the periods 1/4/2018 to 28/2/2020 and 1/11/2021 to 28/2/2022 (PCV13 period). Data from a previous carriage study conducted among healthy and sick children from 1/01/2006 to 30/06/2008 (PCV7 period), were used for comparison of serotype/genotype distributions and antibiotic resistance rates. RESULTS Overall, 76 (19%) children were colonized with S. pneumoniae during the PCV13 period and there were information available from 154 isolates collected during the PCV7 period. Colonization with PCV13 serotypes declined significantly in the PCV13 period compared with historical controls (11% vs 38%, p = 0.0001), being serotypes 19F (8%), 3 (1%) and 6B (1%) the only circulating vaccine types. Serotypes 15B/C and 11A were the most frequently identified non-PCV13 serotypes during the PCV13 period (14% and 11%, respectively); the later one increased significantly between time periods (p = 0.04). Serotype 11A was exclusively associated in the PCV13 period with ampicillin-resistant variants of the Spain9V-ST156 clone (ST6521 and genetically related ST14698), not detected in the preceding period. CONCLUSIONS There was a residual circulation of vaccine types following PCV13 introduction, apart from serotype 19F. Serotype 11A increased between PCV13 and PCV7 periods due to emergence and clonal expansion of ampicillin-resistant genotype ST6521.
Collapse
Affiliation(s)
- Beatriz de Felipe
- Alteraciones Congénitas de la Inmunidad, Instituto de Biomedicina de Sevilla (IBIS), Universidad de Sevilla/Hospital Universitario Virgen del Rocío (HUVR)/CSIC, Sevilla, Spain
| | - Marta Aboza-García
- Alteraciones Congénitas de la Inmunidad, Instituto de Biomedicina de Sevilla (IBIS), Universidad de Sevilla/Hospital Universitario Virgen del Rocío (HUVR)/CSIC, Sevilla, Spain; Centro de Salud Polígono Norte, Sevilla, Spain
| | | | | | | | - Benito Amil-Pérez
- Servicio de Pediatría, Grupo Instituto Hispalense de Pediatría, Sevilla, Spain
| | - Cristóbal Coronel-Rodríguez
- Centro de Salud Amante Laffón, Distrito de Atención Primaria Sevilla, Servicio Andaluz de Salud, Sevilla, Spain
| | | | | | | | - Francisco Javier Morón
- Unidad de Genómica, Instituto de Biomedicina de Sevilla, Universidad de Sevilla/Hospital Universitario Virgen del Rocío/CSIC, Sevilla, Spain
| | - Juan A Cordero-Varela
- Unidad de Bioinformática, Instituto de Biomedicina de Sevilla, Universidad de Sevilla/Hospital Universitario Virgen del Rocío/CSIC, Sevilla, Spain
| | | | - Ignacio Obando
- Alteraciones Congénitas de la Inmunidad, Instituto de Biomedicina de Sevilla (IBIS), Universidad de Sevilla/Hospital Universitario Virgen del Rocío (HUVR)/CSIC, Sevilla, Spain; Sección de Infectología e Inmunopatologías Pediátricas, Hospital Universitario Virgen de Valme, Sevilla, Spain
| |
Collapse
|
18
|
Jansson MK, Nguyen DT, Mikkat S, Warnke C, Janssen MB, Warnke P, Kreikemeyer B, Patenge N. Synthetic mRNA delivered to human cells leads to expression of Cpl-1 bacteriophage-endolysin with activity against Streptococcus pneumoniae. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102145. [PMID: 38435119 PMCID: PMC10907214 DOI: 10.1016/j.omtn.2024.102145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 02/06/2024] [Indexed: 03/05/2024]
Abstract
Endolysins are bacteriophage-encoded hydrolases that show high antibacterial activity and a narrow substrate spectrum. We hypothesize that an mRNA-based approach to endolysin therapy can overcome some challenges of conventional endolysin therapy, namely organ targeting and bioavailability. We show that synthetic mRNA applied to three human cell lines (HEK293T, A549, HepG2 cells) leads to expression and cytosolic accumulation of the Cpl-1 endolysin with activity against Streptococcus pneumoniae. Addition of a human lysozyme signal peptide sequence translocates the Cpl-1 to the endoplasmic reticulum leading to secretion (hlySP-sCpl-1). The pneumococcal killing effect of hlySP-sCpl-1 was enhanced by introduction of a point mutation to avoid N-linked-glycosylation. hlySP-sCpl-1N215D, collected from the culture supernatant of A549 cells 6 h post-transfection showed a significant killing effect and was active against nine pneumococcal strains. mRNA-based cytosolic Cpl-1 and secretory hlySP-sCpl-1N215D show potential for innovative treatment strategies against pneumococcal disease and, to our best knowledge, represent the first approach to mRNA-based endolysin therapy. We assume that many other bacterial pathogens could be targeted with this novel approach.
Collapse
Affiliation(s)
- Moritz K. Jansson
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Dat Tien Nguyen
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Stefan Mikkat
- Core Facility Proteome Analysis, University Medicine Rostock, Rostock, Germany
| | - Carolin Warnke
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Marc Benjamin Janssen
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Philipp Warnke
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Bernd Kreikemeyer
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Nadja Patenge
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| |
Collapse
|
19
|
Soni J, Sinha S, Pandey R. Understanding bacterial pathogenicity: a closer look at the journey of harmful microbes. Front Microbiol 2024; 15:1370818. [PMID: 38444801 PMCID: PMC10912505 DOI: 10.3389/fmicb.2024.1370818] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 02/05/2024] [Indexed: 03/07/2024] Open
Abstract
Bacteria are the most prevalent form of microorganisms and are classified into two categories based on their mode of existence: intracellular and extracellular. While most bacteria are beneficial to human health, others are pathogenic and can cause mild to severe infections. These bacteria use various mechanisms to evade host immunity and cause diseases in humans. The susceptibility of a host to bacterial infection depends on the effectiveness of the immune system, overall health, and genetic factors. Malnutrition, chronic illnesses, and age-related vulnerabilities are the additional confounders to disease severity phenotypes. The impact of bacterial pathogens on public health includes the transmission of these pathogens from healthcare facilities, which contributes to increased morbidity and mortality. To identify the most significant threats to public health, it is crucial to understand the global burden of common bacterial pathogens and their pathogenicity. This knowledge is required to improve immunization rates, improve the effectiveness of vaccines, and consider the impact of antimicrobial resistance when assessing the situation. Many bacteria have developed antimicrobial resistance, which has significant implications for infectious diseases and favors the survival of resilient microorganisms. This review emphasizes the significance of understanding the bacterial pathogens that cause this health threat on a global scale.
Collapse
Affiliation(s)
- Jyoti Soni
- Division of Immunology and Infectious Disease Biology, Integrative Genomics of Host Pathogen Laboratory, Council of Scientific & Industrial Research-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Sristi Sinha
- Division of Immunology and Infectious Disease Biology, Integrative Genomics of Host Pathogen Laboratory, Council of Scientific & Industrial Research-Institute of Genomics and Integrative Biology, New Delhi, India
- School of Biosciences and Technology, Vellore Institute of Technology University, Vellore, India
| | - Rajesh Pandey
- Division of Immunology and Infectious Disease Biology, Integrative Genomics of Host Pathogen Laboratory, Council of Scientific & Industrial Research-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| |
Collapse
|
20
|
Santra S, Nayak I, Paladhi A, Das D, Banerjee A. Estimates of differential toxin expression governing heterogeneous intracellular lifespans of Streptococcus pneumoniae. J Cell Sci 2024; 137:jcs260891. [PMID: 38411297 DOI: 10.1242/jcs.260891] [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: 12/14/2022] [Accepted: 01/10/2024] [Indexed: 02/28/2024] Open
Abstract
Following invasion of the host cell, pore-forming toxins secreted by pathogens compromise vacuole integrity and expose the microbe to diverse intracellular defence mechanisms. However, the quantitative correlation between toxin expression levels and consequent pore dynamics, fostering the intracellular life of pathogens, remains largely unexplored. In this study, using Streptococcus pneumoniae and its secreted pore-forming toxin pneumolysin (Ply) as a model system, we explored various facets of host-pathogen interactions in the host cytosol. Using time-lapse fluorescence imaging, we monitored pore formation dynamics and lifespans of different pneumococcal subpopulations inside host cells. Based on experimental histograms of various event timescales such as pore formation time, vacuolar death or cytosolic escape time and total degradation time, we developed a mathematical model based on first-passage processes that could correlate the event timescales to intravacuolar toxin accumulation. This allowed us to estimate Ply production rate, burst size and threshold Ply quantities that trigger these outcomes. Collectively, we present a general method that illustrates a correlation between toxin expression levels and pore dynamics, dictating intracellular lifespans of pathogens.
Collapse
Affiliation(s)
- Shweta Santra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Indrani Nayak
- Department of Physics, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Ankush Paladhi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Dibyendu Das
- Department of Physics, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Anirban Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| |
Collapse
|
21
|
Fol M, Karpik W, Zablotni A, Kulesza J, Kulesza E, Godkowicz M, Druszczynska M. Innate Lymphoid Cells and Their Role in the Immune Response to Infections. Cells 2024; 13:335. [PMID: 38391948 PMCID: PMC10886880 DOI: 10.3390/cells13040335] [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: 01/05/2024] [Revised: 02/07/2024] [Accepted: 02/10/2024] [Indexed: 02/24/2024] Open
Abstract
Over the past decade, a group of lymphocyte-like cells called innate lymphoid cells (ILCs) has gained considerable attention due to their crucial role in regulating immunity and tissue homeostasis. ILCs, lacking antigen-specific receptors, are a group of functionally differentiated effector cells that act as tissue-resident sentinels against infections. Numerous studies have elucidated the characteristics of ILC subgroups, but the mechanisms controlling protective or pathological responses to pathogens still need to be better understood. This review summarizes the functions of ILCs in the immunology of infections caused by different intracellular and extracellular pathogens and discusses their possible therapeutic potential.
Collapse
Affiliation(s)
- Marek Fol
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
| | - Wojciech Karpik
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
| | - Agnieszka Zablotni
- Department of Bacterial Biology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland;
| | - Jakub Kulesza
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, 91-347 Lodz, Poland;
| | - Ewelina Kulesza
- Department of Rheumatology and Internal Diseases, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Magdalena Godkowicz
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
- Lodz Institutes of the Polish Academy of Sciences, The Bio-Med-Chem Doctoral School, University of Lodz, 90-237 Lodz, Poland
| | - Magdalena Druszczynska
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
| |
Collapse
|
22
|
Wang L, Liu M, Qi Y, Wang J, Shi Q, Xie X, Zhou C, Ma L. hsdSA regulated extracellular vesicle-associated PLY to protect Streptococcus pneumoniae from macrophage killing via LAPosomes. Microbiol Spectr 2024; 12:e0099523. [PMID: 38018988 PMCID: PMC10783081 DOI: 10.1128/spectrum.00995-23] [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: 03/06/2023] [Accepted: 08/01/2023] [Indexed: 11/30/2023] Open
Abstract
IMPORTANCE S. pneumoniae is a major human pathogen that undergoes a spontaneous and reversible phase variation that allows it to survive in different host environments. Interestingly, we found hsdSA , a gene that manipulated the phase variation, promoted the survival and replication of S. pneumoniae in macrophages by regulating EV production and EV-associated PLY. More importantly, here we provided the first evidence that higher EV-associated PLY (produced by D39) could form LAPosomes that were single membrane compartments containing S. pneumoniae, which are induced by integrin β1/NOX2/ROS pathway. At the same time, EV-associated PLY increased the permeability of lysosome membrane and induced an insufficient acidification to escape the host killing, and ultimately prolonged the survival of S. pneumoniae in macrophages. In contrast, lower EV-associated PLY (produced by D39ΔhsdSA ) activated ULK1 recruitment to form double-layered autophagosomes to eliminate bacteria.
Collapse
Affiliation(s)
- Liping Wang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Mengyuan Liu
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yixin Qi
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jian Wang
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Qixue Shi
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xiaolin Xie
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Changlin Zhou
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Lingman Ma
- College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
23
|
Yang S, Chen J, Fu J, Huang J, Li T, Yao Z, Ye X. Disease-Associated Streptococcus pneumoniae Genetic Variation. Emerg Infect Dis 2024; 30:39-49. [PMID: 38146979 PMCID: PMC10756394 DOI: 10.3201/eid3001.221927] [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: 12/27/2023] Open
Abstract
Streptococcus pneumoniae is an opportunistic pathogen that causes substantial illness and death among children worldwide. The genetic backgrounds of pneumococci that cause infection versus asymptomatic carriage vary substantially. To determine the evolutionary mechanisms of opportunistic pathogenicity, we conducted a genomic surveillance study in China. We collected 783 S. pneumoniae isolates from infected and asymptomatic children. By using a 2-stage genomewide association study process, we compared genomic differences between infection and carriage isolates to address genomic variation associated with pathogenicity. We identified 8 consensus k-mers associated with adherence, antimicrobial resistance, and immune modulation, which were unevenly distributed in the infection isolates. Classification accuracy of the best k-mer predictor for S. pneumoniae infection was good, giving a simple target for predicting pathogenic isolates. Our findings suggest that S. pneumoniae pathogenicity is complex and multifactorial, and we provide genetic evidence for precise targeted interventions.
Collapse
|
24
|
Yin F, Hu Y, Bu Z, Liu Y, Zhang H, Hu Y, Xue Y, Li S, Tan C, Chen X, Li L, Zhou R, Huang Q. Genome-wide identification of genes critical for in vivo fitness of multi-drug resistant porcine extraintestinal pathogenic Escherichia coli by transposon-directed insertion site sequencing using a mouse infection model. Virulence 2023; 14:2158708. [PMID: 36537189 PMCID: PMC9828833 DOI: 10.1080/21505594.2022.2158708] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) is an important zoonotic pathogen. Recently, ExPEC has been reported to be an emerging problem in pig farming. However, the mechanism of pathogenicity of porcine ExPEC remains to be revealed. In this study, we constructed a transposon (Tn) mutagenesis library covering Tn insertion in over 72% of the chromosome-encoded genes of a virulent and multi-drug resistant porcine ExPEC strain PCN033. By using a mouse infection model, a transposon-directed insertion site sequencing (TraDIS) assay was performed to identify in vivo fitness factors. By comparing the Tn insertion frequencies between the input Tn library and the recovered library from different organs, 64 genes were identified to be involved in fitness during systemic infection. 15 genes were selected and individual gene deletion mutants were constructed. The in vivo fitness was evaluated by using a competitive infection assay. Among them, ΔfimG was significantly outcompeted by the WT strain in vivo and showed defective adhesion to host cells. rfa which was involved in lipopolysaccharide biosynthesis was shown to be critical for in vivo fitness which may have resulted from its role in the resistance to serum killing. In addition, several metabolic genes including fepB, sdhC, fepG, gltS, dcuA, ccmH, ddpD, narU, glpD, malM, and yabL and two regulatory genes metJ and baeS were shown as important determinants of in vivo fitness of porcine ExPEC. Collectively, this study performed a genome-wide screening for in vivo fitness factors which will be important for understanding the pathogenicity of porcine ExPEC.
Collapse
Affiliation(s)
- Fan Yin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yan Hu
- College of Animal Sciences & Technology, Huazhong Agricultural University, Wuhan, China
| | - Zixuan Bu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yuying Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Hui Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yawen Hu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ying Xue
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shaowen Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chen Tan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China,Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China,Ministry of Science and Technology, International Research Center for Animal Disease, Wuhan, China
| | - Xiabing Chen
- Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Science and Technology, Wuhan, China
| | - Lu Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China,Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China,Ministry of Science and Technology, International Research Center for Animal Disease, Wuhan, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China,Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China,Ministry of Science and Technology, International Research Center for Animal Disease, Wuhan, China,The HZAU-HVSEN Institute, Wuhan, China,CONTACT Rui Zhou
| | - Qi Huang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China,Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China,Ministry of Science and Technology, International Research Center for Animal Disease, Wuhan, China,Qi Huang
| |
Collapse
|
25
|
Zou Y, Wang H, Fang J, Sun H, Deng X, Wang J, Deng Y, Chi G. Isorhamnetin as a novel inhibitor of pneumolysin against Streptococcus pneumoniae infection in vivo/in vitro. Microb Pathog 2023; 185:106382. [PMID: 37839759 DOI: 10.1016/j.micpath.2023.106382] [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: 04/10/2023] [Revised: 09/18/2023] [Accepted: 10/05/2023] [Indexed: 10/17/2023]
Abstract
The increasing incidence of Streptococcus pneumoniae (S. pneumoniae) infection severely threatened the global public heath, causing a significant fatality in immunocompromised hosts. Notably, pneumolysin (PLY) as a pore-forming cytolysin plays a crucial role in the pathogenesis of pneumococcal pneumonia and lung injury. In this study, a natural flavonoid isorhamnetin was identified as a PLY inhibition to suppress PLY-induced hemolysis by engaging the predicted residues and attenuate cytolysin PLY-mediated A549 cells injury. Underlying mechanisms revealed that PLY inhibitor isorhamnetin further contributed to decrease the formation of bacterial biofilms without affecting the expression of PLY. In vivo S. pneumoniae infection confirmed that the pathological injury of lung tissue evoked by S. pneumoniae was ameliorated by isorhamnetin treatment. Collectively, these results presented that isorhamnetin could inhibit the biological activity of PLY, thus reducing the pathogenicity of S. pneumoniae. In summary, our study laid a foundation for the feasible anti-virulence strategy targeting PLY, and provided a promising PLY inhibitor for the treatment of S. pneumoniae infection.
Collapse
Affiliation(s)
- Yinuo Zou
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Haiting Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Juan Fang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hongxiang Sun
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Xuming Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianfeng Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yanhong Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Gefu Chi
- The Affiliated Hospital of Inner Mongolia Medical University, Huhhot, Nei Monggol, China.
| |
Collapse
|
26
|
Obolski U, Swarthout TD, Kalizang'oma A, Mwalukomo TS, Chan JM, Weight CM, Brown C, Cave R, Cornick J, Kamng'ona AW, Msefula J, Ercoli G, Brown JS, Lourenço J, Maiden MC, French N, Gupta S, Heyderman RS. The metabolic, virulence and antimicrobial resistance profiles of colonising Streptococcus pneumoniae shift after PCV13 introduction in urban Malawi. Nat Commun 2023; 14:7477. [PMID: 37978177 PMCID: PMC10656543 DOI: 10.1038/s41467-023-43160-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/02/2023] [Indexed: 11/19/2023] Open
Abstract
Streptococcus pneumoniae causes substantial mortality among children under 5-years-old worldwide. Polysaccharide conjugate vaccines (PCVs) are highly effective at reducing vaccine serotype disease, but emergence of non-vaccine serotypes and persistent nasopharyngeal carriage threaten this success. We investigated the hypothesis that following vaccine, adapted pneumococcal genotypes emerge with the potential for vaccine escape. We genome sequenced 2804 penumococcal isolates, collected 4-8 years after introduction of PCV13 in Blantyre, Malawi. We developed a pipeline to cluster the pneumococcal population based on metabolic core genes into "Metabolic genotypes" (MTs). We show that S. pneumoniae population genetics are characterised by emergence of MTs with distinct virulence and antimicrobial resistance (AMR) profiles. Preliminary in vitro and murine experiments revealed that representative isolates from emerging MTs differed in growth, haemolytic, epithelial infection, and murine colonisation characteristics. Our results suggest that in the context of PCV13 introduction, pneumococcal population dynamics had shifted, a phenomenon that could further undermine vaccine control and promote spread of AMR.
Collapse
Affiliation(s)
- Uri Obolski
- Department of Epidemiology and Preventive Medicine, School of Public Health, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Porter School of the Environment and Earth Sciences, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel.
| | - Todd D Swarthout
- Malawi Liverpool Wellcome Programme, Blantyre, Malawi
- Mucosal Pathogens Research Group, Research Department of Infection, Division of Infection & Immunity, University College London, London, United Kingdom
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Akuzike Kalizang'oma
- Malawi Liverpool Wellcome Programme, Blantyre, Malawi
- Mucosal Pathogens Research Group, Research Department of Infection, Division of Infection & Immunity, University College London, London, United Kingdom
| | | | - Jia Mun Chan
- Mucosal Pathogens Research Group, Research Department of Infection, Division of Infection & Immunity, University College London, London, United Kingdom
| | - Caroline M Weight
- Mucosal Pathogens Research Group, Research Department of Infection, Division of Infection & Immunity, University College London, London, United Kingdom
- Faculty of Health and Medicine, Biomedical and Life Sciences, Lancaster University, Lancaster, United Kingdom
- Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| | - Comfort Brown
- Malawi Liverpool Wellcome Programme, Blantyre, Malawi
| | - Rory Cave
- Mucosal Pathogens Research Group, Research Department of Infection, Division of Infection & Immunity, University College London, London, United Kingdom
| | - Jen Cornick
- Malawi Liverpool Wellcome Programme, Blantyre, Malawi
- Clinical Infection, Microbiology and Immunology, Institute of Infection Veterinary & Ecological Science, University of Liverpool, Liverpool, United Kingdom
| | | | | | - Giuseppe Ercoli
- UCL Respiratory, Division of Medicine, University College London, London, United Kingdom
| | - Jeremy S Brown
- UCL Respiratory, Division of Medicine, University College London, London, United Kingdom
| | - José Lourenço
- Department of Zoology, University of Oxford, Oxford, United Kingdom
- Universidade Católica Portuguesa, Faculty of Medicine, Biomedical Research Centre, Lisbon, Portugal
| | - Martin C Maiden
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Neil French
- Clinical Infection, Microbiology and Immunology, Institute of Infection Veterinary & Ecological Science, University of Liverpool, Liverpool, United Kingdom
| | - Sunetra Gupta
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Robert S Heyderman
- Malawi Liverpool Wellcome Programme, Blantyre, Malawi.
- Mucosal Pathogens Research Group, Research Department of Infection, Division of Infection & Immunity, University College London, London, United Kingdom.
| |
Collapse
|
27
|
Williams AN, Ma A, Croxen MA, Demczuk WHB, Martin I, Tyrrell GJ. Genomic analysis of Streptococcus pneumoniae serogroup 20 isolates in Alberta, Canada from 1993-2019. Microb Genom 2023; 9:001141. [PMID: 38015202 PMCID: PMC10711305 DOI: 10.1099/mgen.0.001141] [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: 07/17/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023] Open
Abstract
In the province of Alberta, Canada, invasive disease caused by Streptococcus pneumoniae serogroup 20 (serotypes 20A/20B) has been increasing in incidence. Here, we characterize provincial invasive serogroup 20 isolates collected from 1993 to 2019 alongside invasive and non-invasive serogroup 20 isolates from the Global Pneumococcal Sequencing (GPS) Project collected from 1998 to 2015. Trends in clinical metadata and geographic location were evaluated, and serogroup 20 isolate genomes were subjected to molecular sequence typing, virulence and antimicrobial resistance factor mining, phylogenetic analysis and pangenome calculation. Two hundred and seventy-four serogroup 20 isolates from Alberta were sequenced, and analysed along with 95 GPS Project genomes. The majority of invasive Alberta serogroup 20 isolates were identified after 2007 in primarily middle-aged adults and typed predominantly as ST235, a sequence type that was rare among GPS Project isolates. Most Alberta isolates carried a full-length whaF capsular gene, suggestive of serotype 20B. All Alberta and GPS Project genomes carried molecular resistance determinants implicated in fluoroquinolone and macrolide resistance, with a few Alberta isolates exhibiting phenotypic resistance to azithromycin, clindamycin, erythromycin, tetracycline and trimethoprim-sulfamethoxazole, as well as non-susceptibility to tigecycline. All isolates carried multiple virulence factors including those involved in adherence, immune modulation and nutrient uptake, as well as exotoxins and exoenzymes. Phylogenetically, Alberta serogroup 20 isolates clustered with predominantly invasive GPS Project isolates from the USA, Israel, Brazil and Nepal. Overall, this study highlights the increasing incidence of invasive S. pneumoniae serogroup 20 disease in Alberta, Canada, and provides insights into the genetic and clinical characteristics of these isolates within a global context.
Collapse
Affiliation(s)
- Ashley N. Williams
- Department of Laboratory Medicine and Pathology, University of Alberta, 5-411 Edmonton Clinic Health Academy, Edmonton, Alberta, T6G 1C9 Canada
- Alberta Precision Laboratory – Public Health Laboratory, 8440 112 Street NW, Edmonton, Alberta, T6G 2B7 Canada
| | - Angela Ma
- Department of Laboratory Medicine and Pathology, University of Alberta, 5-411 Edmonton Clinic Health Academy, Edmonton, Alberta, T6G 1C9 Canada
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, Ste. #1100, Salt Lake City, UT, 84112 USA
| | - Matthew A. Croxen
- Department of Laboratory Medicine and Pathology, University of Alberta, 5-411 Edmonton Clinic Health Academy, Edmonton, Alberta, T6G 1C9 Canada
- Alberta Precision Laboratory – Public Health Laboratory, 8440 112 Street NW, Edmonton, Alberta, T6G 2B7 Canada
- Li Ka Shing Institute of Virology, University of Alberta, 6-010 Katz Centre for Health Research, 11315 - 87 Ave NW, Edmonton, Alberta, T6G 2E1 Canada
- Women & Children’s Health Research Institute (WCHRI), University of Alberta, 5-083 Edmonton Clinic Health Academy (ECHA), 11405 87 Avenue NW Edmonton, AB, T6G 1C9 Canada
| | - Walter H. B. Demczuk
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, R3E 3R2 Canada
| | - Irene Martin
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, R3E 3R2 Canada
| | - Gregory J. Tyrrell
- Department of Laboratory Medicine and Pathology, University of Alberta, 5-411 Edmonton Clinic Health Academy, Edmonton, Alberta, T6G 1C9 Canada
- Alberta Precision Laboratory – Public Health Laboratory, 8440 112 Street NW, Edmonton, Alberta, T6G 2B7 Canada
- Li Ka Shing Institute of Virology, University of Alberta, 6-010 Katz Centre for Health Research, 11315 - 87 Ave NW, Edmonton, Alberta, T6G 2E1 Canada
| |
Collapse
|
28
|
Zahari NIN, Engku Abd Rahman ENS, Irekeola AA, Ahmed N, Rabaan AA, Alotaibi J, Alqahtani SA, Halawi MY, Alamri IA, Almogbel MS, Alfaraj AH, Ibrahim FA, Almaghaslah M, Alissa M, Yean CY. A Review of the Resistance Mechanisms for β-Lactams, Macrolides and Fluoroquinolones among Streptococcus pneumoniae. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1927. [PMID: 38003976 PMCID: PMC10672801 DOI: 10.3390/medicina59111927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/22/2023] [Accepted: 10/29/2023] [Indexed: 11/26/2023]
Abstract
Streptococcus pneumoniae (S. pneumoniae) is a bacterial species often associated with the occurrence of community-acquired pneumonia (CAP). CAP refers to a specific kind of pneumonia that occurs in individuals who acquire the infection outside of a healthcare setting. It represents the leading cause of both death and morbidity on a global scale. Moreover, the declaration of S. pneumoniae as one of the 12 leading pathogens was made by the World Health Organization (WHO) in 2017. Antibiotics like β-lactams, macrolides, and fluoroquinolones are the primary classes of antimicrobial medicines used for the treatment of S. pneumoniae infections. Nevertheless, the efficacy of these antibiotics is diminishing as a result of the establishment of resistance in S. pneumoniae against these antimicrobial agents. In 2019, the WHO declared that antibiotic resistance was among the top 10 hazards to worldwide health. It is believed that penicillin-binding protein genetic alteration causes β-lactam antibiotic resistance. Ribosomal target site alterations and active efflux pumps cause macrolide resistance. Numerous factors, including the accumulation of mutations, enhanced efflux mechanisms, and plasmid gene acquisition, cause fluoroquinolone resistance. Furthermore, despite the advancements in pneumococcal vaccinations and artificial intelligence (AI), it is not feasible for individuals to rely on them indefinitely. The ongoing development of AI for combating antimicrobial resistance necessitates more research and development efforts. A few strategies can be performed to curb this resistance issue, including providing educational initiatives and guidelines, conducting surveillance, and establishing new antibiotics targeting another part of the bacteria. Hence, understanding the resistance mechanism of S. pneumoniae may aid researchers in developing a more efficacious antibiotic in future endeavors.
Collapse
Affiliation(s)
- Nurul Izzaty Najwa Zahari
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian 16150, Malaysia (E.N.S.E.A.R.)
| | - Engku Nur Syafirah Engku Abd Rahman
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian 16150, Malaysia (E.N.S.E.A.R.)
| | - Ahmad Adebayo Irekeola
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian 16150, Malaysia (E.N.S.E.A.R.)
- Microbiology Unit, Department of Biological Sciences, College of Natural and Applied Sciences, Summit University Offa, Offa PMB 4412, Nigeria
| | - Naveed Ahmed
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian 16150, Malaysia (E.N.S.E.A.R.)
| | - Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Jawaher Alotaibi
- Infectious Diseases Unit, Department of Medicine, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | | | - Mohammed Y. Halawi
- Cytogenetics Department, Dammam Regional Laboratory and Blood Bank, Dammam 31411, Saudi Arabia
| | - Ibrahim Ateeq Alamri
- Blood Bank Department, Dammam Regional Laboratory and Blood Bank, Dammam 31411, Saudi Arabia
| | - Mohammed S. Almogbel
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail 4030, Saudi Arabia
| | - Amal H. Alfaraj
- Pediatric Department, Abqaiq General Hospital, First Eastern Health Cluster, Abqaiq 33261, Saudi Arabia
| | - Fatimah Al Ibrahim
- Infectious Disease Division, Department of Internal Medicine, Dammam Medical Complex, Dammam 32245, Saudi Arabia
| | - Manar Almaghaslah
- Infectious Disease Division, Department of Internal Medicine, Dammam Medical Complex, Dammam 32245, Saudi Arabia
| | - Mohammed Alissa
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Chan Yean Yean
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian 16150, Malaysia (E.N.S.E.A.R.)
- Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Health Campus, Kubang Kerian 16150, Malaysia
| |
Collapse
|
29
|
Lobb B, Lee MC, McElheny CL, Doi Y, Yahner K, Hoberman A, Martin JM, Hirota JA, Doxey AC, Shaikh N. Genomic classification and antimicrobial resistance profiling of Streptococcus pneumoniae and Haemophilus influenza isolates associated with paediatric otitis media and upper respiratory infection. BMC Infect Dis 2023; 23:596. [PMID: 37700242 PMCID: PMC10498559 DOI: 10.1186/s12879-023-08560-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/24/2023] [Indexed: 09/14/2023] Open
Abstract
Acute otitis media (AOM) is the most common childhood bacterial infectious disease requiring antimicrobial therapy. Most cases of AOM are caused by translocation of Streptococcus pneumoniae or Haemophilus influenzae from the nasopharynx to the middle ear during an upper respiratory tract infection (URI). Ongoing genomic surveillance of these pathogens is important for vaccine design and tracking of emerging variants, as well as for monitoring patterns of antibiotic resistance to inform treatment strategies and stewardship.In this work, we examined the ability of a genomics-based workflow to determine microbiological and clinically relevant information from cultured bacterial isolates obtained from patients with AOM or an URI. We performed whole genome sequencing (WGS) and analysis of 148 bacterial isolates cultured from the nasopharynx (N = 124, 94 AOM and 30 URI) and ear (N = 24, all AOM) of 101 children aged 6-35 months presenting with AOM or an URI. We then performed WGS-based sequence typing and antimicrobial resistance profiling of each strain and compared results to those obtained from traditional microbiological phenotyping.WGS of clinical isolates resulted in 71 S. pneumoniae genomes and 76 H. influenzae genomes. Multilocus sequencing typing (MSLT) identified 33 sequence types for S. pneumoniae and 19 predicted serotypes including the most frequent serotypes 35B and 3. Genome analysis predicted 30% of S. pneumoniae isolates to have complete or intermediate penicillin resistance. AMR predictions for S. pneumoniae isolates had strong agreement with clinical susceptibility testing results for beta-lactam and non beta-lactam antibiotics, with a mean sensitivity of 93% (86-100%) and a mean specificity of 98% (94-100%). MLST identified 29 H. influenzae sequence types. Genome analysis identified beta-lactamase genes in 30% of H. influenzae strains, which was 100% in agreement with clinical beta-lactamase testing. We also identified a divergent highly antibiotic-resistant strain of S. pneumoniae, and found its closest sequenced strains, also isolated from nasopharyngeal samples from over 15 years ago.Ultimately, our work provides the groundwork for clinical WGS-based workflows to aid in detection and analysis of H. influenzae and S. pneumoniae isolates.
Collapse
Affiliation(s)
- Briallen Lobb
- Department of Biology and Waterloo Centre for Microbial Research, University of Waterloo, Waterloo, ON, Canada
| | - Matthew C Lee
- University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Division of General Academic Pediatrics, Pittsburgh, USA
| | - Christi L McElheny
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yohei Doi
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kristin Yahner
- University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Division of General Academic Pediatrics, Pittsburgh, USA
| | - Alejandro Hoberman
- University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Division of General Academic Pediatrics, Pittsburgh, USA
| | - Judith M Martin
- University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Division of General Academic Pediatrics, Pittsburgh, USA
| | - Jeremy A Hirota
- Department of Biology and Waterloo Centre for Microbial Research, University of Waterloo, Waterloo, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Hospital, Hamilton, ON, Canada
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Andrew C Doxey
- Department of Biology and Waterloo Centre for Microbial Research, University of Waterloo, Waterloo, ON, Canada.
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
| | - Nader Shaikh
- University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Division of General Academic Pediatrics, Pittsburgh, USA.
| |
Collapse
|
30
|
Hernandez-Morfa M, Olivero NB, Zappia VE, Piñas GE, Reinoso-Vizcaino NM, Cian MB, Nuñez-Fernandez M, Cortes PR, Echenique J. The oxidative stress response of Streptococcus pneumoniae: its contribution to both extracellular and intracellular survival. Front Microbiol 2023; 14:1269843. [PMID: 37789846 PMCID: PMC10543277 DOI: 10.3389/fmicb.2023.1269843] [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: 07/30/2023] [Accepted: 08/28/2023] [Indexed: 10/05/2023] Open
Abstract
Streptococcus pneumoniae is a gram-positive, aerotolerant bacterium that naturally colonizes the human nasopharynx, but also causes invasive infections and is a major cause of morbidity and mortality worldwide. This pathogen produces high levels of H2O2 to eliminate other microorganisms that belong to the microbiota of the respiratory tract. However, it also induces an oxidative stress response to survive under this stressful condition. Furthermore, this self-defense mechanism is advantageous in tolerating oxidative stress imposed by the host's immune response. This review provides a comprehensive overview of the strategies employed by the pneumococcus to survive oxidative stress. These strategies encompass the utilization of H2O2 scavengers and thioredoxins, the adaptive response to antimicrobial host oxidants, the regulation of manganese and iron homeostasis, and the intricate regulatory networks that control the stress response. Here, we have also summarized less explored aspects such as the involvement of reparation systems and polyamine metabolism. A particular emphasis is put on the role of the oxidative stress response during the transient intracellular life of Streptococcus pneumoniae, including coinfection with influenza A and the induction of antibiotic persistence in host cells.
Collapse
Affiliation(s)
- Mirelys Hernandez-Morfa
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nadia B. Olivero
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Victoria E. Zappia
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - German E. Piñas
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nicolas M. Reinoso-Vizcaino
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Melina B. Cian
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mariana Nuñez-Fernandez
- Centro de Química Aplicada, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Paulo R. Cortes
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Jose Echenique
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
31
|
Graham AS, Ben-Azu B, Tremblay MÈ, Torre P, Senekal M, Laughton B, van der Kouwe A, Jankiewicz M, Kaba M, Holmes MJ. A review of the auditory-gut-brain axis. Front Neurosci 2023; 17:1183694. [PMID: 37600010 PMCID: PMC10435389 DOI: 10.3389/fnins.2023.1183694] [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: 03/10/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Hearing loss places a substantial burden on medical resources across the world and impacts quality of life for those affected. Further, it can occur peripherally and/or centrally. With many possible causes of hearing loss, there is scope for investigating the underlying mechanisms involved. Various signaling pathways connecting gut microbes and the brain (the gut-brain axis) have been identified and well established in a variety of diseases and disorders. However, the role of these pathways in providing links to other parts of the body has not been explored in much depth. Therefore, the aim of this review is to explore potential underlying mechanisms that connect the auditory system to the gut-brain axis. Using select keywords in PubMed, and additional hand-searching in google scholar, relevant studies were identified. In this review we summarize the key players in the auditory-gut-brain axis under four subheadings: anatomical, extracellular, immune and dietary. Firstly, we identify important anatomical structures in the auditory-gut-brain axis, particularly highlighting a direct connection provided by the vagus nerve. Leading on from this we discuss several extracellular signaling pathways which might connect the ear, gut and brain. A link is established between inflammatory responses in the ear and gut microbiome-altering interventions, highlighting a contribution of the immune system. Finally, we discuss the contribution of diet to the auditory-gut-brain axis. Based on the reviewed literature, we propose numerous possible key players connecting the auditory system to the gut-brain axis. In the future, a more thorough investigation of these key players in animal models and human research may provide insight and assist in developing effective interventions for treating hearing loss.
Collapse
Affiliation(s)
- Amy S. Graham
- Imaging Sciences, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Human Biology, Division of Biomedical Engineering, University of Cape Town, Cape Town, South Africa
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec City, QC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Quebec City, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Peter Torre
- School of Speech, Language, and Hearing Sciences, San Diego State University, San Diego, CA, United States
| | - Marjanne Senekal
- Department of Human Biology, Division of Physiological Sciences, University of Cape Town, Cape Town, South Africa
| | - Barbara Laughton
- Family Clinical Research Unit, Department of Pediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
| | - Andre van der Kouwe
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, United States
- Department of Radiology, Harvard Medical School, Boston, MA, United States
| | - Marcin Jankiewicz
- Imaging Sciences, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Human Biology, Division of Biomedical Engineering, University of Cape Town, Cape Town, South Africa
| | - Mamadou Kaba
- Department of Pathology, Division of Medical Microbiology, University of Cape Town, Cape Town, South Africa
| | - Martha J. Holmes
- Imaging Sciences, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Human Biology, Division of Biomedical Engineering, University of Cape Town, Cape Town, South Africa
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- ImageTech, Simon Fraser University, Surrey, BC, Canada
| |
Collapse
|
32
|
Ong HH, Toh WK, Thong LY, Phoon LQ, Clarke SC, Cheah ESG. Investigation of Upper Respiratory Carriage of Bacterial Pathogens among University Students in Kampar, Malaysia. Trop Med Infect Dis 2023; 8:tropicalmed8050269. [PMID: 37235317 DOI: 10.3390/tropicalmed8050269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
The carriage of bacterial pathogens in the human upper respiratory tract (URT) is associated with a risk of invasive respiratory tract infections, but the related epidemiological information on this at the population level is scarce in Malaysia. This study aimed to investigate the URT carriage of Streptococcus pneumoniae, Haemophilus influenzae, Neisseria meningitidis, Staphylococcus aureus, Klebsiella pneumoniae and Pseudomonas aeruginosa among 100 university students by nasal and oropharyngeal swabbing. The presence of S. aureus, K. pneumoniae and P. aeruginosa was assessed via swab culture on selective media and PCR on the resulting isolates. For S. pneumoniae, H. influenzae and N. meningitidis, their presence was assessed via multiplex PCR on the total DNA extracts from chocolate agar cultures. The carriage prevalence of H. influenzae, S. aureus, S. pneumoniae, K. pneumoniae, N. meningitidis and P. aeruginosa among the subjects was 36%, 27%, 15%, 11%, 5% and 1%, respectively, by these approaches. Their carriage was significantly higher in males compared to females overall. The S. aureus, K. pneumoniae and P. aeruginosa isolates were also screened by the Kirby-Bauer assay, in which 51.6% of S. aureus were penicillin-resistant. The outcomes from carriage studies are expected to contribute to informing infectious disease control policies and guidelines.
Collapse
Affiliation(s)
- Hing Huat Ong
- Department of Biological Science, Faculty of Science, Kampar Campus, Universiti Tunku Abdul Rahman, Kampar 31900, Malaysia
| | - Wai Keat Toh
- Department of Biological Science, Faculty of Science, Kampar Campus, Universiti Tunku Abdul Rahman, Kampar 31900, Malaysia
| | - Li Ying Thong
- Department of Biological Science, Faculty of Science, Kampar Campus, Universiti Tunku Abdul Rahman, Kampar 31900, Malaysia
| | - Lee Quen Phoon
- Department of Allied Health Sciences, Faculty of Science, Kampar Campus, Universiti Tunku Abdul Rahman, Kampar 31900, Malaysia
- Centre for Biomedical and Nutrition Research, Kampar Campus, Universiti Tunku Abdul Rahman, Kampar 31900, Malaysia
| | - Stuart C Clarke
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Trust, Southampton SO16 6YD, UK
- Global Health Research Institute, University of Southampton, Southampton SO17 1BJ, UK
- School of Postgraduate Studies, International Medical University, Kuala Lumpur 57000, Malaysia
- Centre for Translational Research, Institute for Research, Development, and Innovation, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Eddy Seong Guan Cheah
- Department of Biological Science, Faculty of Science, Kampar Campus, Universiti Tunku Abdul Rahman, Kampar 31900, Malaysia
- Centre for Biomedical and Nutrition Research, Kampar Campus, Universiti Tunku Abdul Rahman, Kampar 31900, Malaysia
| |
Collapse
|
33
|
Han X, Meng X, Wang X, Leng S, Liu Q, Zhang L, Li P, Zhang Q, Hu HY. Analyte-Triggered Excited-State Intramolecular Proton Transfer- Delayed Fluorescence: A General Approach for Time-Resolved Turn-On Fluorescence Imaging. Anal Chem 2023; 95:7715-7722. [PMID: 37125992 DOI: 10.1021/acs.analchem.3c00827] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The research of delayed fluorescence (DF) has been a hot topic in biological imaging. However, the development of analyte-triggered small molecule DF probes remains a considerable challenge. Herein a novel excited-state intramolecular proton transfer-delayed fluorescence (ESIPT-DF) approach to construct analyte-stimulated DF probes was reported. These new classes of ESIPT-DF luminophores were strategically designed and synthesized by incorporating 2-(2'-hydroxyphenyl)benzothiazole (HBT), a known ESIPT-based fluorophore, as acceptor with a series of classic donor moieties, which formed a correspondingly twisted donor-acceptor pair within each molecule. Thereinto, HBT-PXZ and HBT-PTZ exhibited significant ESIPT and DF characters with lifetimes of 5.37 and 3.65 μs in the solid state, respectively. Furthermore, a caged probe HBT-PXZ-Ga was developed by introducing a hydrophilic d-galactose group as the recognition unit specific for β-galactosidase (β-gal) and ESIPT-DF blocking agent and applied to investigate the influence of metal ions on β-gal activity on the surface of Streptococcus pneumoniae as a convenient tool. This ESIPT-DF "turn-on" approach is easily adaptable for the measurement of many different analytes using only a predictable modification on the caged group without modification of the core structure.
Collapse
Affiliation(s)
- Xiaowan Han
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiangchuan Meng
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiang Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Shan Leng
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Qian Liu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Leilei Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Pingping Li
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Qingyang Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hai-Yu Hu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
34
|
Leal JT, Primon-Barros M, de Carvalho Robaina A, Pizzutti K, Mott MP, Trentin DS, Dias CAG. Streptococcus pneumoniae serotype 19A from carriers and invasive disease: virulence gene profile and pathogenicity in a Galleria mellonella model. Eur J Clin Microbiol Infect Dis 2023; 42:399-411. [PMID: 36790530 DOI: 10.1007/s10096-023-04560-6] [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: 07/26/2022] [Accepted: 01/27/2023] [Indexed: 02/16/2023]
Abstract
PURPOSE This study aimed to evaluate and compare the presence of genes related to surface proteins between isolates of Streptococcus pneumoniae from healthy carriers (HC) and invasive pneumococcal disease (IPD) with a particular focus on serotype 19A. METHODS The presence of these genes was identified by real-time PCR. Subsequently, we employed the Galleria mellonella larval infection model to study their effect on pathogenicity in vivo. RESULTS The percentage of selected virulence genes was similar between the HC and IPD groups (p > 0.05), and the genes lytA, nanB, pavA, pcpA, phtA, phtB, phtE, rrgA, and sipA were all present in both groups. However, the virulence profile of the isolates differed individually between HC and IPD groups. The highest lethality in G. mellonella was for IPD isolates (p < 0.01), even when the virulence profile was the same as compared to the HC isolates or when the nanA, pspA, pspA-fam1, and pspC genes were not present. CONCLUSIONS The occurrence of the investigated virulence genes was similar between HC and IPD S. pneumoniae serotype 19A groups. However, the IPD isolates showed a higher lethality in the alternative G. mellonella model than the HC isolates, regardless of the virulence gene composition, indicating that other virulence factors may play a decisive role in virulence. Currently, this is the first report using the in vivo G. mellonella model to study the virulence of clinical isolates of S. pneumoniae.
Collapse
Affiliation(s)
- Josiane Trevisol Leal
- Laboratório de Microbiologia Molecular, Departamento de Ciências Básicas da Saúde, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande Do Sul, Brasil
- Laboratório de Bacteriologia & Modelos Experimentais Alternativos, Departamento de Ciências Básicas da Saúde, Programa de Pós-Graduação em Biociências (PPGBIO), Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brasil
| | - Muriel Primon-Barros
- Laboratório de Microbiologia Molecular, Departamento de Ciências Básicas da Saúde, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande Do Sul, Brasil
| | - Amanda de Carvalho Robaina
- Laboratório de Microbiologia Molecular, Departamento de Ciências Básicas da Saúde, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande Do Sul, Brasil
| | - Kauana Pizzutti
- Laboratório de Microbiologia Molecular, Departamento de Ciências Básicas da Saúde, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande Do Sul, Brasil
| | - Mariana Preussler Mott
- Laboratório de Microbiologia Molecular, Departamento de Ciências Básicas da Saúde, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande Do Sul, Brasil
| | - Danielle Silva Trentin
- Laboratório de Bacteriologia & Modelos Experimentais Alternativos, Departamento de Ciências Básicas da Saúde, Programa de Pós-Graduação em Biociências (PPGBIO), Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brasil.
| | - Cícero Armídio Gomes Dias
- Laboratório de Microbiologia Molecular, Departamento de Ciências Básicas da Saúde, Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande Do Sul, Brasil
| |
Collapse
|
35
|
Chun YY, Tan KS, Yu L, Pang M, Wong MHM, Nakamoto R, Chua WZ, Huee-Ping Wong A, Lew ZZR, Ong HH, Chow VT, Tran T, Yun Wang D, Sham LT. Influence of glycan structure on the colonization of Streptococcus pneumoniae on human respiratory epithelial cells. Proc Natl Acad Sci U S A 2023; 120:e2213584120. [PMID: 36943879 PMCID: PMC10068763 DOI: 10.1073/pnas.2213584120] [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: 08/08/2022] [Accepted: 02/10/2023] [Indexed: 03/23/2023] Open
Abstract
Virtually all living cells are encased in glycans. They perform key cellular functions such as immunomodulation and cell-cell recognition. Yet, how their composition and configuration affect their functions remains enigmatic. Here, we constructed isogenic capsule-switch mutants harboring 84 types of capsular polysaccharides (CPSs) in Streptococcus pneumoniae. This collection enables us to systematically measure the affinity of structurally related CPSs to primary human nasal and bronchial epithelial cells. Contrary to the paradigm, the surface charge does not appreciably affect epithelial cell binding. Factors that affect adhesion to respiratory cells include the number of rhamnose residues and the presence of human-like glycomotifs in CPS. Besides, pneumococcal colonization stimulated the production of interleukin 6 (IL-6), granulocyte-macrophage colony-stimulating factor (GM-CSF), and monocyte chemoattractantprotein-1 (MCP-1) in nasal epithelial cells, which also appears to be dependent on the serotype. Together, our results reveal glycomotifs of surface polysaccharides that are likely to be important for colonization and survival in the human airway.
Collapse
Affiliation(s)
- Ye-Yu Chun
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
| | - Kai Sen Tan
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117597
| | - Lisa Yu
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- College of Art and Sciences, Cornell University, Ithaca, NY14853
| | - Michelle Pang
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
| | - Ming Hui Millie Wong
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
| | - Rei Nakamoto
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
| | - Wan-Zhen Chua
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
| | - Amanda Huee-Ping Wong
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117593
| | - Zhe Zhang Ryan Lew
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
| | - Hsiao Hui Ong
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
| | - Vincent T. Chow
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
| | - Thai Tran
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117593
| | - De Yun Wang
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
| | - Lok-To Sham
- Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117545
| |
Collapse
|
36
|
CALABRÒ GIOVANNAELISA, VITALE FRANCESCO, RIZZO CATERINA, PUGLIESE ANDREA, BOCCALINI SARA, BECHINI ANGELA, PANATTO DONATELLA, AMICIZIA DANIELA, DOMNICH ALEXANDER, AMODIO EMANUELE, COSTANTINO CLAUDIO, DI PIETRO MARIALUISA, SALVATI CRISTINA, D’AMBROSIO FLORIANA, ORSINI FRANCESCA, MAIDA ADA, DOMINICI ANNA, CLEMENTE DANIA, CECCI MARINA, PELLACCHIA ANDREA, DI SERAFINO FRANCESCA, BAKKER KEVIN, MALIK TUFAILMOHAMMAD, SHAROMI OLUWASEUN, BELLUZZO MIRIAM, LEONFORTE FRANCESCO, ZAGRA LUIGI, LA GATTA EMANUELE, PETRELLA LUIGI, BONANNI PAOLO, DE WAURE CHIARA. [The new 15-valent pneumococcal conjugate vaccine for the prevention of S. pneumoniae infections in pediatric age: a Health Technology Assessment]. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2023; 64:E1-E160. [PMID: 37655211 PMCID: PMC10468156 DOI: 10.15167/2421-4248/jpmh2023.64.1s1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Affiliation(s)
- GIOVANNA ELISA CALABRÒ
- Sezione di Igiene, Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italia
- VIHTALI (Value In Health Technology and Academy for Leadership & Innovation), Spin-off dell’Università Cattolica del Sacro Cuore, Roma, Italia
| | - FRANCESCO VITALE
- Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, Università degli Studi di Palermo
| | - CATERINA RIZZO
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università degli Studi di Pisa, Pisa, Italia
| | - ANDREA PUGLIESE
- Dipartimento di Matematica, Università di Trento, Trento, Italia
| | - SARA BOCCALINI
- Dipartimento di Scienze della Salute, Università degli di Studi di Firenze, Firenze, Italia
| | - ANGELA BECHINI
- Dipartimento di Scienze della Salute, Università degli di Studi di Firenze, Firenze, Italia
| | - DONATELLA PANATTO
- Dipartimento di Scienze della Salute, Università degli Studi di Genova, Genova, Italia
- Centro Interuniversitario di Ricerca sull’Influenza e le altre Infezioni Trasmissibili (CIRI-IT), Genova, Italia
| | - DANIELA AMICIZIA
- Dipartimento di Scienze della Salute, Università degli Studi di Genova, Genova, Italia
- Centro Interuniversitario di Ricerca sull’Influenza e le altre Infezioni Trasmissibili (CIRI-IT), Genova, Italia
| | | | - EMANUELE AMODIO
- Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, Università degli Studi di Palermo
| | - CLAUDIO COSTANTINO
- Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, Università degli Studi di Palermo
| | - MARIA LUISA DI PIETRO
- Sezione di Igiene, Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - CRISTINA SALVATI
- Dipartimento di Scienze della Salute, Università degli di Studi di Firenze, Firenze, Italia
| | - FLORIANA D’AMBROSIO
- Sezione di Igiene, Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - FRANCESCA ORSINI
- Alta Scuola di Economia e Management dei Sistemi Sanitari (ALTEMS), Università Cattolica del Sacro Cuore, Roma, Italia
| | - ADA MAIDA
- Sezione di Igiene, Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - ANNA DOMINICI
- Dipartimento di Medicina e Chirurgia, Università degli di Studi di Perugia, Italia
| | - DANIA CLEMENTE
- Dipartimento di Medicina e Chirurgia, Università degli di Studi di Perugia, Italia
| | - MARINA CECCI
- Dipartimento di Medicina e Chirurgia, Università degli di Studi di Perugia, Italia
| | - ANDREA PELLACCHIA
- Dipartimento di Medicina e Chirurgia, Università degli di Studi di Perugia, Italia
| | - FRANCESCA DI SERAFINO
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università degli Studi di Pisa, Pisa, Italia
| | - KEVIN BAKKER
- Health Economic and Decision Sciences (HEDS), Biostatistics & Research Decision Sciences (BARDS), Merck Research Laboratories, West Point, Pennsylvania
| | - TUFAIL MOHAMMAD MALIK
- Health Economic and Decision Sciences (HEDS), Biostatistics & Research Decision Sciences (BARDS), Merck Research Laboratories, West Point, Pennsylvania
| | - OLUWASEUN SHAROMI
- Health Economic and Decision Sciences (HEDS), Biostatistics & Research Decision Sciences (BARDS), Merck Research Laboratories, West Point, Pennsylvania
| | - MIRIAM BELLUZZO
- Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, Università degli Studi di Palermo
| | - FRANCESCO LEONFORTE
- Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, Università degli Studi di Palermo
| | - LUIGI ZAGRA
- Dipartimento di Promozione della Salute, Materno-Infantile, di Medicina Interna e Specialistica di Eccellenza “G. D’Alessandro”, Università degli Studi di Palermo
| | - EMANUELE LA GATTA
- Sezione di Igiene, Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - LUIGI PETRELLA
- Sezione di Igiene, Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - PAOLO BONANNI
- Dipartimento di Scienze della Salute, Università degli di Studi di Firenze, Firenze, Italia
| | - CHIARA DE WAURE
- Dipartimento di Medicina e Chirurgia, Università degli di Studi di Perugia, Italia
| |
Collapse
|
37
|
Fritsch VN, Linzner N, Busche T, Said N, Weise C, Kalinowski J, Wahl MC, Antelmann H. The MerR-family regulator NmlR is involved in the defense against oxidative stress in Streptococcus pneumoniae. Mol Microbiol 2023; 119:191-207. [PMID: 36349475 DOI: 10.1111/mmi.14999] [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: 07/15/2022] [Revised: 10/25/2022] [Accepted: 10/30/2022] [Indexed: 11/11/2022]
Abstract
Streptococcus pneumoniae has to cope with the strong oxidant hypochlorous acid (HOCl), during host-pathogen interactions. Thus, we analyzed the global gene expression profile of S. pneumoniae D39 towards HOCl stress. In the RNA-seq transcriptome, the NmlR, SifR, CtsR, HrcA, SczA and CopY regulons and the etrx1-ccdA1-msrAB2 operon were most strongly induced under HOCl stress, which participate in the oxidative, electrophile and metal stress response in S. pneumoniae. The MerR-family regulator NmlR harbors a conserved Cys52 and controls the alcohol dehydrogenase-encoding adhC gene under carbonyl and NO stress. We demonstrated that NmlR senses also HOCl stress to activate transcription of the nmlR-adhC operon. HOCl-induced transcription of adhC required Cys52 of NmlR in vivo. Using mass spectrometry, NmlR was shown to be oxidized to intersubunit disulfides or S-glutathionylated under oxidative stress in vitro. A broccoli-FLAP-based assay further showed that both NmlR disulfides significantly increased transcription initiation at the nmlR promoter by RNAP in vitro, which depends on Cys52. Phenotype analyses revealed that NmlR functions in the defense against oxidative stress and promotes survival of S. pneumoniae during macrophage infections. In conclusion, NmlR was characterized as HOCl-sensing transcriptional regulator, which activates transcription of adhC under oxidative stress by thiol switches in S. pneumoniae.
Collapse
Affiliation(s)
| | - Nico Linzner
- Institute of Biology-Microbiology, Freie Universität Berlin, Berlin, Germany
| | - Tobias Busche
- Center for Biotechnology, University Bielefeld, Bielefeld, Germany.,NGS Core Facility, Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Nelly Said
- Laboratory of Structural Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Christoph Weise
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Jörn Kalinowski
- Center for Biotechnology, University Bielefeld, Bielefeld, Germany
| | - Markus C Wahl
- Laboratory of Structural Biochemistry, Freie Universität Berlin, Berlin, Germany.,Macromolecular Crystallography, Helmholtz-Zentrum Berlin für Materialien und Energie, Berlin, Germany
| | - Haike Antelmann
- Institute of Biology-Microbiology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
38
|
Minhas V, Domenech A, Synefiaridou D, Straume D, Brendel M, Cebrero G, Liu X, Costa C, Baldry M, Sirard JC, Perez C, Gisch N, Hammerschmidt S, Håvarstein LS, Veening JW. Competence remodels the pneumococcal cell wall exposing key surface virulence factors that mediate increased host adherence. PLoS Biol 2023; 21:e3001990. [PMID: 36716340 PMCID: PMC9910801 DOI: 10.1371/journal.pbio.3001990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 02/09/2023] [Accepted: 01/04/2023] [Indexed: 02/01/2023] Open
Abstract
Competence development in the human pathogen Streptococcus pneumoniae controls several features such as genetic transformation, biofilm formation, and virulence. Competent bacteria produce so-called "fratricins" such as CbpD that kill noncompetent siblings by cleaving peptidoglycan (PGN). CbpD is a choline-binding protein (CBP) that binds to phosphorylcholine residues found on wall and lipoteichoic acids (WTA and LTA) that together with PGN are major constituents of the pneumococcal cell wall. Competent pneumococci are protected against fratricide by producing the immunity protein ComM. How competence and fratricide contribute to virulence is unknown. Here, using a genome-wide CRISPRi-seq screen, we show that genes involved in teichoic acid (TA) biosynthesis are essential during competence. We demonstrate that LytR is the major enzyme mediating the final step in WTA formation, and that, together with ComM, is essential for immunity against CbpD. Importantly, we show that key virulence factors PspA and PspC become more surface-exposed at midcell during competence, in a CbpD-dependent manner. Together, our work supports a model in which activation of competence is crucial for host adherence by increased surface exposure of its various CBPs.
Collapse
Affiliation(s)
- Vikrant Minhas
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, Lausanne, Switzerland
| | - Arnau Domenech
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, Lausanne, Switzerland
| | - Dimitra Synefiaridou
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, Lausanne, Switzerland
| | - Daniel Straume
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Max Brendel
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, Universität Greifswald, Greifswald, Germany
| | | | - Xue Liu
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, Lausanne, Switzerland,Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Charlotte Costa
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Mara Baldry
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Jean-Claude Sirard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Camilo Perez
- Biozentrum, University of Basel, Basel, Switzerland
| | - Nicolas Gisch
- Division of Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, Universität Greifswald, Greifswald, Germany
| | - Leiv Sigve Håvarstein
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway,* E-mail: (LSH); (J-WV)
| | - Jan-Willem Veening
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, Lausanne, Switzerland,* E-mail: (LSH); (J-WV)
| |
Collapse
|
39
|
Wang C, Yang Y, Cao Y, Liu K, Shi H, Guo X, Liu W, Hao R, Song H, Zhao R. Nanocarriers for the delivery of antibiotics into cells against intracellular bacterial infection. Biomater Sci 2023; 11:432-444. [PMID: 36503914 DOI: 10.1039/d2bm01489k] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The barrier function of host cells enables intracellular bacteria to evade the lethality of the host immune system and antibiotics, thereby causing chronic and recurrent infections that seriously threaten human health. Currently, the main clinical strategy for the treatment of intracellular bacterial infections involves the use of long-term and high-dose antibiotics. However, insufficient intracellular delivery of antibiotics along with various resistance mechanisms not only weakens the efficacy of current therapies but also causes serious adverse drug reactions, further increasing the disease and economic burden. Improving the delivery efficiency, intracellular accumulation, and action time of antibiotics remains the most economical and effective way to treat intracellular bacterial infections. The rapid development of nanotechnology provides a strategy to efficiently deliver antibiotics against intracellular bacterial infections into cells. In this review, we summarize the types of common intracellular pathogens, the difficulties faced by antibiotics in the treatment of intracellular bacterial infections, and the research progress of several types of representative nanocarriers for the delivery of antibiotics against intracellular bacterial infections that have emerged in recent years. This review is expected to provide a reference for further elucidating the intracellular transport mechanism of nanocarrier-drug complexes, designing safer and more effective nanocarriers and establishing new strategies against intracellular bacterial infection.
Collapse
Affiliation(s)
- Chao Wang
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Yi Yang
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Yuanyuan Cao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Kaixin Liu
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Hua Shi
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Xudong Guo
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Wanying Liu
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Rongzhang Hao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Hongbin Song
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Rongtao Zhao
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| |
Collapse
|
40
|
de Felipe B, Aboza-García M, González-Galán V, Salamanca de la Cueva I, Martín-Quintero JA, Amil-Pérez B, Coronel-Rodríguez C, Palacios-Soria MÁ, García Ruiz-Santaquiteria MI, Torres-Sánchez MJ, Morón FJ, Cordero-Varela JA, Obando-Pacheco P, Obando I. Epidemiología molecular de la colonización nasofaríngea neumocócica en niños de Sevilla, tras la implementación del programa de vacunación con VNC13 en Andalucía (España). Enferm Infecc Microbiol Clin 2023. [DOI: 10.1016/j.eimc.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
41
|
Suo W, Guo X, Zhang X, Xiao S, Wang S, Yin Y, Zheng Y. Glucose levels affect MgaSpn regulation on the virulence and adaptability of Streptococcus pneumoniae. Microb Pathog 2023; 174:105896. [PMID: 36460142 DOI: 10.1016/j.micpath.2022.105896] [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: 09/20/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022]
Abstract
Streptococcus pneumoniae can regulate virulence gene expression by sensing environmental changes, which is key to its pathogenicity. The global transcription regulator MgaSpn of Streptococcus pneumoniae regulates virulence genes expression by directly binding to the promoter regions, but its role in response to different environments remains unclear. In this study, we found that glucose levels could affect phosphocholine content, which was mediated by MgaSpn. MgaSpn can also alter its anti-phagocytosis ability, depending on the availability of glucose. In addition, transcriptome analysis of wild-type D39s in low and high glucose concentrations revealed that MgaSpn was also involved in the regulation of carbon metabolism inhibition (carbon catabolite repression; CCR) and translation processes, which made S. pneumoniae highly competitive in fluctuating environments. In conclusion, MgaSpn is closely related to the virulence and environmental adaptability of Streptococcus pneumoniae.
Collapse
Affiliation(s)
- Weicai Suo
- Department of Medicine Laboratory, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, and Chongqing Key Laboratory of Pediatrics, Chongqing, PR China
| | - Xinlin Guo
- Department of Medicine Laboratory, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, and Chongqing Key Laboratory of Pediatrics, Chongqing, PR China
| | - Xuemei Zhang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, PR China
| | - Shengnan Xiao
- Precision Medicine Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Shuhui Wang
- Department of Medicine Laboratory, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, and Chongqing Key Laboratory of Pediatrics, Chongqing, PR China
| | - Yibing Yin
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, PR China
| | - Yuqiang Zheng
- Department of Medicine Laboratory, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, and Chongqing Key Laboratory of Pediatrics, Chongqing, PR China.
| |
Collapse
|
42
|
Therapeutic potential of kaempferol on Streptococcus pneumoniae infection. Microbes Infect 2023; 25:105058. [PMID: 36216303 PMCID: PMC9540706 DOI: 10.1016/j.micinf.2022.105058] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022]
Abstract
Co-infections with pathogens and secondary bacterial infections play significant roles during the pandemic coronavirus disease 2019 (COVID-19) pathogenetic process, caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Notably, co-infections with Streptococcus pneumoniae (S. pneumoniae), as a major Gram-positive pathogen causing pneumonia or meningitis, severely threaten the diagnosis, therapy, and prognosis of COVID-19 worldwide. Accumulating evidences have emerged indicating that S. pneumoniae evolves multiple virulence factors, including pneumolysin (PLY) and sortase A (SrtA), which have been extensively explored as alternative anti-infection targets. In our study, natural flavonoid kaempferol was identified as a potential candidate drug for infection therapeutics via anti-virulence mechanisms. We found that kaempferol could interfere with the pore-forming activity of PLY by engaging with catalytic active sites and consequently inhibit PLY-mediated cytotoxicity. Additionally, exposed to kaempferol significantly reduced the SrtA peptidase activity by occupying the active sites of SrtA. Further, the biofilms formation and bacterial adhesion to the host cells could be significantly thwarted by kaempferol incubation. In vivo infection model by S. pneumoniae highlighted that kaempferol oral administration exhibited notable treatment benefits, as evidenced by decreased bacterial burden, suggesting that kaempferol has tremendous potential to attenuate S. pneumoniae pathogenicity. Scientifically, our study implies that kaempferol is a promising therapeutic option by targeting bacterial virulence factors.
Collapse
|
43
|
Geerts N, De Vooght L, Passaris I, Delputte P, Van den Bergh B, Cos P. Antibiotic Tolerance Indicative of Persistence Is Pervasive among Clinical Streptococcus pneumoniae Isolates and Shows Strong Condition Dependence. Microbiol Spectr 2022; 10:e0270122. [PMID: 36374111 PMCID: PMC9769776 DOI: 10.1128/spectrum.02701-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 10/16/2022] [Indexed: 11/16/2022] Open
Abstract
Streptococcus pneumoniae is an important human pathogen, being one of the most common causes of community-acquired pneumonia and otitis media. Antibiotic resistance in S. pneumoniae is an emerging problem, as it depletes our arsenal of effective drugs. In addition, persistence also contributes to the antibiotic crisis in many other pathogens, yet for S. pneumoniae, little is known about antibiotic-tolerant persisters and robust experimental means are lacking. Persister cells are phenotypic variants that exist as a subpopulation within a clonal culture. Being tolerant to lethal antibiotics, they underly the chronic nature of a variety of infections and even help in acquiring genetic resistance. In this study, we set out to identify and characterize persistence in S. pneumoniae. Specifically, we followed different strategies to overcome the self-limiting nature of S. pneumoniae as a confounding factor in the prolonged monitoring of antibiotic survival needed to study persistence. Under optimized conditions, we identified genuine persisters in various growth phases and for four relevant antibiotics through biphasic survival dynamics and heritability assays. Finally, we detected a high variety in antibiotic survival levels across a diverse collection of S. pneumoniae clinical isolates, which assumes that a high natural diversity in persistence is widely present in S. pneumoniae. Collectively, this proof of concept significantly progresses the understanding of the importance of antibiotic persistence in S. pneumoniae infections, which will set the stage for characterizing its relevance to clinical outcomes and advocates for increased attention to the phenotype in both fundamental and clinical research. IMPORTANCE S. pneumoniae is considered a serious threat by the Centers for Disease Control and Prevention because of rising antibiotic resistance. In addition to resistance, bacteria can also survive lethal antibiotic treatment by developing antibiotic tolerance, more specifically, antibiotic tolerance through persistence. This phenotypic variation seems omnipresent among bacterial life, is linked to therapy failure, and acts as a catalyst for resistance development. This study gives the first proof of the presence of persister cells in S. pneumoniae and shows a high variety in persistence levels among diverse strains, suggesting that persistence is a general trait in S. pneumoniae cultures. Our work advocates for higher interest for persistence in S. pneumoniae as a contributing factor for therapy failure and resistance development.
Collapse
Affiliation(s)
- Nele Geerts
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Linda De Vooght
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | | | - Peter Delputte
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| | - Bram Van den Bergh
- Centre of Microbial and Plant Genetics, Department of Molecular and Microbial Systems, KU Leuven, Leuven, Belgium
- Center for Microbiology, Flanders Institute for Biotechnology, VIB, Leuven, Belgium
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), Wilrijk, Belgium
| |
Collapse
|
44
|
Gao N, Rezaee F. Airway Epithelial Cell Junctions as Targets for Pathogens and Antimicrobial Therapy. Pharmaceutics 2022; 14:2619. [PMID: 36559113 PMCID: PMC9786141 DOI: 10.3390/pharmaceutics14122619] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Intercellular contacts between epithelial cells are established and maintained by the apical junctional complexes (AJCs). AJCs conserve cell polarity and build epithelial barriers to pathogens, inhaled allergens, and environmental particles in the respiratory tract. AJCs consist of tight junctions (TJs) and adherens junctions (AJs), which play a key role in maintaining the integrity of the airway barrier. Emerging evidence has shown that different microorganisms cause airway barrier dysfunction by targeting TJ and AJ proteins. This review discusses the pathophysiologic mechanisms by which several microorganisms (bacteria and viruses) lead to the disruption of AJCs in airway epithelial cells. We present recent progress in understanding signaling pathways involved in the formation and regulation of cell junctions. We also summarize the potential chemical inhibitors and pharmacological approaches to restore the integrity of the airway epithelial barrier. Understanding the AJCs-pathogen interactions and mechanisms by which microorganisms target the AJC and impair barrier function may further help design therapeutic innovations to treat these infections.
Collapse
Affiliation(s)
- Nannan Gao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Fariba Rezaee
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
- Center for Pediatric Pulmonary Medicine, Cleveland Clinic Children’s, Cleveland, OH 44195, USA
| |
Collapse
|
45
|
Characterization of Streptococcus pneumoniae Macrolide Resistance and Its Mechanism in Northeast China over a 20-Year Period. Microbiol Spectr 2022; 10:e0054622. [PMID: 35938873 PMCID: PMC9602527 DOI: 10.1128/spectrum.00546-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Due to the resistance of Streptococcus pneumoniae to β-lactams, macrolides, and tetracyclines, treatment alternatives have become increasingly limited worldwide. We aim to describe the characterization of erythromycin-resistant S. pneumoniae (ERSP) strains in northeastern China over a period of 20 years. A total of 1,240 ERSP strains were collected and classified into five groups based on the ages of the patients. Etest strips and Kirby-Bauer disk diffusion were performed for drug susceptibility testing. The capsule swelling test was used for capsule typing. The phenotype of drug resistance was detected by the erythromycin and clindamycin double-disk method. The ermB, ermTR, mefA, and tetM genes were detected by PCR. Among the 1,240 ERSP strains, 510 were invasive isolates, and 730 were noninvasive isolates. The results of drug susceptibility testing showed that the rates of resistance to penicillin, amoxicillin, cefotaxime, ceftriaxone, meropenem, tetracycline, trimethoprim-sulfamethoxazole, and chloramphenicol varied among the different age groups. 19F, 19A, 23F, 14, and 6B were the serotypes that were commonly found among ERSP strains. Among all strains, 99.03% (1,228/1,240) exhibited an MLSB (macrolide-lincosamide-streptogramin B) resistance phenotype, of which 1,221 strains displayed a constitutive MLSB (cMLSB) phenotype and 7 strains showed an inducible MLSB (iMLSB) phenotype. All of these strains carried the ermB gene. In contrast, only 0.97% of strains of M phenotypes were found to carry the mefA gene. Both the ermB and mefA genes were detected in 704 strains that exhibited multidrug resistance, whereas the ermTR gene was not detected. Furthermore, 1,185 tetracycline-resistant strains were found to carry the tetM gene. Macrolide antimicrobial drugs should be used cautiously for the empirical treatment of S. pneumoniae infections. IMPORTANCE This study presents a retrospective analysis using 1,240 clinical erythromycin-resistant Streptococcus pneumoniae (ERSP) isolates collected in northeastern China between January 2000 and December 2019. The serotype distribution, corresponding vaccine coverage, as well as resistance phenotypes, genes, and mechanisms to macrolide and tetracycline of these isolates were systematically described, analyzed, and discussed. We hope that this study will inform clinicians in their respective regions when selecting antimicrobial agents. We also hope that this study is useful for researchers in related fields. Finally, we emphasize in this study that vaccination is the best preventive measure for S. pneumoniae infection considering its resistance to commonly used antibiotics. The determination of the S. pneumoniae serotype distribution also provides valuable empirical evidence for local health authorities when introducing appropriate vaccines in a specific area.
Collapse
|
46
|
Alexandrova AS, Pencheva DR, Setchanova LP, Gergova RT. Association of pili with widespread multidrug-resistant genetic lineages of non-invasive pediatric Streptococcus pneumoniae isolates. Acta Microbiol Immunol Hung 2022; 69:177-184. [PMID: 36094859 DOI: 10.1556/030.2022.01816] [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: 07/01/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022]
Abstract
The study aimed to evaluate the presence of pili in non-invasive pediatric pneumococcal isolates and to elucidate possible links with genetic lineages, serotypes, and antimicrobial resistance. We examined 147 Streptococcus pneumoniae isolates from children with respiratory tract infections and acute otitis media. Serotyping was performed by latex agglutination and capsule swelling reaction. Serogroup 6 was subjected to PCR-serotyping. Minimum inhibitory concentrations were determined according to EUCAST breakpoints. PCRs for rlrA and pitB genes were performed to detect a presence of type 1 and type 2 pili. MLST was conducted to define the clonal structure of the piliated strains. Almost all children (96.5%) were vaccinated with the pneumococcal conjugate vaccine PCV10. We detected 76.8% non-PCV10 - serotypes (NVTs) and 14.3% PCV10 serotypes. The predominant serotypes were NVTs: 19A (14.3%), 6C (12.2%), 3 (9.5%), 15A (7.5%) and 6A (6.8%). PI-1 was detected among 10.9% non-PCV10 serotypes 6A, 6C, and 19A and 6.1% PCV10 serotypes 19F and 23F. Type 2 pili were not found in the studied population. High levels of antimicrobial nonsusceptibility to erythromycin (58.5%), oral penicillin (55.8%), clindamycin (46.9%), trimethoprim-sulfamethoxazole (45.6%), tetracycline (39.5%) and ceftriaxone (16.3%) were revealed. The multidrug-resistant strains (MDR) were 55.1%. MLST represented 18 STs and three CCs among the piliated pneumococci: CC386, CC320, and CC81. More than half of the piliated strains (56.0%) belonged to successfully circulating international clones. PI-1 was associated mainly with MDR 6A, 6C, 19A, 19F, and 23F isolates from the widespread CC386, CC320, and CC81.
Collapse
Affiliation(s)
- Alexandra Sashova Alexandrova
- 1 Department of Medical Microbiology, Medical Faculty, Medical University of Sofia, 1, G.Sofiiski Boul., 1431-Sofia, Bulgaria
| | - Daniela Rosenova Pencheva
- 2 Department of Medical Chemistry and Biochemistry, Molecular Medicine Center, Medical Faculty, Medical University of Sofia, 2, Zdrave str., 1431-Sofia, Bulgaria
| | - Lena Petrova Setchanova
- 1 Department of Medical Microbiology, Medical Faculty, Medical University of Sofia, 1, G.Sofiiski Boul., 1431-Sofia, Bulgaria
| | - Raina Tsvetanova Gergova
- 1 Department of Medical Microbiology, Medical Faculty, Medical University of Sofia, 1, G.Sofiiski Boul., 1431-Sofia, Bulgaria
| |
Collapse
|
47
|
Safdarpour S, Eftekhari Z, Eidi A, Doroud D. Encapsulated saponin by ferritin nanoparticles attenuates the murine pneumococcal pneumonia. Microb Pathog 2022; 172:105731. [PMID: 36041710 DOI: 10.1016/j.micpath.2022.105731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/03/2022] [Accepted: 08/16/2022] [Indexed: 10/31/2022]
Abstract
Streptococcus pneumonia is classified as a gram-positive bacterial pathogen that causes asymptomatic or symptomatic respiratory infections. This study aimed to evaluate the effects of designed encapsulated saponin by ferritin nanoparticles in the healing progression of experimental bacterial pneumonia. The saponin encapsulated by ferritin followed the disassembly-reassembly process. Pneumonia was induced by the preparation of Streptococcus pneumonia. A total of 50 NMRI mice were divided into control, pneumonia, pneumonia + ferritin, pneumonia + saponin, and pneumonia + encapsulated saponin by ferritin nanoparticles (Nano Saponin) groups. ELISA, Real-time PCR, and Western blotting were used to measure sera IL-4 level, tumor necrosis factor-alpha (Tnf-α), and protein cyclooxygenase-2 (COX-2) gene expression, respectively. COX-2 protein expression, Tnf-α gene expression, and serum levels of IL-4 reduced compared to the pneumonia group. The histopathology results revealed that the rates of inflammation, mucus secretion, pulmonary hemorrhage, thickening of the alveoli wall, and secretion of inflammatory cells were lower in the Nano Saponin group than in the other groups. This study suggests that Glycyrrhiza glabra saponin and encapsulated saponin by ferritin nanoparticles oral consumption with anti-Tnf-α effect besides decreasing protein expression of COX-2 allows mice with pneumonia to recover.
Collapse
Affiliation(s)
- Saba Safdarpour
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zohre Eftekhari
- Quality Control Department, Research & Production Complex, Pasteur Institute of Iran, Alborz, Iran.
| | - Akram Eidi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Delaram Doroud
- Research & Production Complex, Pasteur Institute of Iran, Alborz, Iran
| |
Collapse
|
48
|
Meredith JD, Chapman I, Ulrich K, Sebastian C, Stull F, Gray MJ. Escherichia coli RclA is a highly active hypothiocyanite reductase. Proc Natl Acad Sci U S A 2022; 119:e2119368119. [PMID: 35867824 PMCID: PMC9335216 DOI: 10.1073/pnas.2119368119] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 05/20/2022] [Indexed: 01/24/2023] Open
Abstract
Hypothiocyanite and hypothiocyanous acid (OSCN-/HOSCN) are pseudohypohalous acids released by the innate immune system which are capable of rapidly oxidizing sulfur-containing amino acids, causing significant protein aggregation and damage to invading bacteria. HOSCN is abundant in saliva and airway secretions and has long been considered a highly specific antimicrobial that is nearly harmless to mammalian cells. However, certain bacteria, commensal and pathogenic, are able to escape damage by HOSCN and other harmful antimicrobials during inflammation, which allows them to continue to grow and, in some cases, cause severe disease. The exact genes or mechanisms by which bacteria respond to HOSCN have not yet been elucidated. We have found, in Escherichia coli, that the flavoprotein RclA, previously implicated in reactive chlorine resistance, reduces HOSCN to thiocyanate with near-perfect catalytic efficiency and strongly protects E. coli against HOSCN toxicity. This is notable in E. coli because this species thrives in the chronically inflamed environment found in patients with inflammatory bowel disease and is able to compete with and outgrow other important commensal organisms, suggesting that HOSCN may be a relevant antimicrobial in the gut, which has not previously been explored. RclA is conserved in a variety of epithelium-colonizing bacteria, implicating its HOSCN reductase activity in a variety of host-microbe interactions. We show that an rclA mutant of the probiotic Limosilactobacillus reuteri is sensitive to HOSCN and that RclA homologs from Staphylococcus aureus, Streptococcus pneumoniae, and Bacteroides thetaiotaomicron all have potent protective activity against HOSCN when expressed in E. coli.
Collapse
Affiliation(s)
- Julia D. Meredith
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233
| | - Irina Chapman
- Department of Chemistry, Western Michigan University, Kalamazoo, MI 49008
| | - Kathrin Ulrich
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Caitlyn Sebastian
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233
| | - Frederick Stull
- Department of Chemistry, Western Michigan University, Kalamazoo, MI 49008
| | - Michael J. Gray
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233
| |
Collapse
|
49
|
Huang LD, Yang MJ, Huang YY, Jiang KY, Yan J, Sun AH. Molecular Characterization of Predominant Serotypes, Drug Resistance, and Virulence Genes of Streptococcus pneumoniae Isolates From East China. Front Microbiol 2022; 13:892364. [PMID: 35722327 PMCID: PMC9198556 DOI: 10.3389/fmicb.2022.892364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Streptococcus pneumoniae is a common diplococcus pathogen found worldwide. The characterization of predominant serotypes, drug resistance, and virulence genes of S. pneumoniae isolates prevailing in different areas and countries is clinically important for choice of antibiotics and improvement of vaccines. In this study, pneumonia (78.7%) and meningitis (37.0%) were the predominant diseases observed in the 282 (children) and 27 (adults) S. pneumoniae-infected patients (p < 0.05) from seven hospitals in different areas of East China. Of the 309 pneumococcal isolates, 90.3% were classified by PCR into 15 serotypes, with serotypes 19F (27.2%) and the 6A/B (19.1%) being most predominant (p < 0.05). Importantly, serotypes 15A and 15B/C combined for a total of 10.4% of the isolates, but these serotypes are not included in the 13-valent pneumococcal capsule conjugate vaccine used in China. Antimicrobial susceptibility analysis by the E-test showed that >95% of the 309 pneumococcal isolates were susceptible to moxifloxacin and levofloxacin, as well as 18.4, 85.8, and 81.6% of the isolates displayed susceptibility to penicillin, cefotaxime, and imipenem, respectively. A significant correlation between the prevalence of predominant serotypes and their penicillin resistance was observed (p < 0.05). In particular, >95% of all the pneumococcal isolates showed resistance to erythromycin and azithromycin. Of the nine detected virulence genes, the lytA, ply, hysA, and nanA were the most common with 95–100% positive rates in the 309 pneumococcal isolates, while the pavA and psaA genes displayed a significant correlation with pneumococcal bacteremia and meningitis (p < 0.05). Overall, our data suggested that the predominant serotypes, drug resistance, and virulence genes of the S. pneumoniae isolates prevailing in East China are distinct from those observed in other areas of China and adjacent countries.
Collapse
Affiliation(s)
- Li-Dan Huang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Mei-Juan Yang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Yan-Ying Huang
- Hangzhou Chest Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke-Yi Jiang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Jie Yan
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, China
| | - Ai-Hua Sun
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
50
|
Membrane particles evoke a serotype-independent cross-protection against pneumococcal infection that is dependent on the conserved lipoproteins MalX and PrsA. Proc Natl Acad Sci U S A 2022; 119:e2122386119. [PMID: 35648835 PMCID: PMC9191655 DOI: 10.1073/pnas.2122386119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
SignificancePneumococcal infections are major contributors to morbidity and mortality worldwide. Introduction of pneumococcal conjugated vaccines (PCVs) into the childhood vaccination program has led to a decrease in invasive pneumococcal disease (IPD) in vaccinated children but concurrently to an increase of nonvaccine-type IPD, also in nonvaccinated age groups such as the elderly. Thus, novel vaccine approaches are urgently needed, especially for the elderly, targeting all pneumococci causing IPD. Here, we show that pneumococcal membrane particles (MPs) evoke a serotype-independent cross-protection against IPD. This protection is dependent on the presence of the two conserved lipoproteins MalX and PrsA. We suggest that MPs can be used for pneumococcal vaccine development.
Collapse
|