1
|
Narciso AR, Dookie R, Nannapaneni P, Normark S, Henriques-Normark B. Streptococcus pneumoniae epidemiology, pathogenesis and control. Nat Rev Microbiol 2025; 23:256-271. [PMID: 39506137 DOI: 10.1038/s41579-024-01116-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 11/08/2024]
Abstract
Infections caused by Streptococcus pneumoniae (also known as pneumococci) pose a threat to human health. Pneumococcal infections are the most common cause of milder respiratory tract infections, such as otitis and sinusitis, and of more severe diseases, including pneumonia (with or without septicaemia) and meningitis. The introduction of pneumococcal conjugate vaccines in the childhood vaccination programme in many countries has led to a notable decrease of severe invasive pneumococcal disease in vaccinated children. However, infections caused by non-vaccine types have concurrently increased, causing invasive pneumococcal disease in unvaccinated populations (such as older adults), which has hampered the effect of these vaccines. Moreover, emerging antibiotic resistance is threatening effective therapy. Thus, new approaches are needed for the treatment and prevention of pneumococcal infections, and recent advances in the field may pave the way for new strategies. Recently, several important findings have been gained regarding pneumococcal epidemiology, genomics and the effect of the introduction of pneumococcal conjugate vaccines and of the COVID-19 pandemic. Moreover, elucidative pathogenesis studies have shown that the interactions between pneumococcal virulence factors and host receptors may be exploited for new therapies, and new vaccine candidates have been suggested. In this Review, we summarize some recent findings from clinical disease to basic pathogenesis studies that may be of importance for future control strategies.
Collapse
Affiliation(s)
- Ana Rita Narciso
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Rebecca Dookie
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Priyanka Nannapaneni
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Staffan Normark
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Birgitta Henriques-Normark
- Department of Microbiology, Tumour and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
2
|
Sun H, Li X, Yang X, Qin J, Liu Y, Zheng Y, Wang Q, Liu R, Sun H, Chen X, Zhang Q, Jia T, Wu X, Feng L, Wang L, Liu B. Low leucine levels in the blood enhance the pathogenicity of neonatal meningitis-causing Escherichia coli. Nat Commun 2025; 16:2466. [PMID: 40075077 PMCID: PMC11904087 DOI: 10.1038/s41467-025-57850-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Neonatal bacterial meningitis is associated with substantial mortality and morbidity worldwide. Neonatal meningitis-causing Escherichia coli (NMEC) is the most common gram-negative bacteria responsible for this disease. However, the interactions of NMEC with its environment within the host are poorly understood. Here, we showed that a low level of leucine, a niche-specific signal in the blood, promotes NMEC pathogenicity by enhancing bacterial survival and replication in the blood. A low leucine level downregulates the expression of NsrP, a small RNA (sRNA) identified in this study, in NMEC in an Lrp-dependent manner. NsrP destabilizes the mRNA of the purine biosynthesis-related gene purD by direct base pairing. Decreased NsrP expression in response to low leucine levels in the blood, which is a purine-limiting environment, activates the bacterial de novo purine biosynthesis pathway, thereby enhancing bacterial pathogenicity in the host. Deletion of NsrP or purD significantly increases or decreases the development of E. coli bacteremia and meningitis in animal models, respectively. Furthermore, we showed that intravenous administration of leucine effectively reduces the development of bacteremia and meningitis caused by NMEC by blocking the Lrp-NsrP-PurD signal transduction pathway. This study provides a potential strategy for the prevention and treatment of E. coli-induced meningitis.
Collapse
Affiliation(s)
- Hao Sun
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Xiaoya Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Xinyuan Yang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Jingliang Qin
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Yutao Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Yangyang Zheng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Qian Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Ruiying Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Hongmin Sun
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Xintong Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Qiyue Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Tianyuan Jia
- Shenzhen National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Xiaoxue Wu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Lu Feng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China
- Nankai International Advanced Research Institute, Shenzhen, China
| | - Lei Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China.
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China.
- Southwest United Graduate School, Kunming, 650092, China.
| | - Bin Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, 300457, China.
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, 300071, China.
- Nankai International Advanced Research Institute, Shenzhen, China.
| |
Collapse
|
3
|
Tran VTA, Zhu X, Jamsranjav A, Lee LP, Cho H. Escherichia Coli K1-colibactin meningitis induces microglial NLRP3/IL-18 exacerbating H3K4me3-synucleinopathy in human inflammatory gut-brain axis. Commun Biol 2025; 8:382. [PMID: 40050667 PMCID: PMC11885818 DOI: 10.1038/s42003-025-07787-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
Escherichia coli K1 (E. coli K1) meningitis early occurs in the gastrointestinal and causes severe damage to the central nervous system, including lifelong neurological complications in survivors. However, the cellular mechanism by which E. coli K1 may cause neuropathies is not well understood due to the lack of relevant human multi-organ models for studying multifaceted systemic inflammation across the gut-brain axis. Here, we reconstruct a multicellular model of the human gut-brain axis to identify the neuropathogenic mechanism driven by E. coli K1-colibactin meningitis. We observed that E. coli K1-genotoxic colibactin induced intestinal and peripheral interleukin 6, causing the blood-brain barrier injury and endothelial inflammation via the p38/p65 pathways. Serpin-E1 from the damaged cerebral endothelia induces reactive astrocytes to release IFN-γ, which reduces microglial phagocytosis of E. coli K1 and exacerbates detrimental neuroinflammation via NLRP3/IL-18 axis. Microglial IL-18 elevates neuronal reactive oxidative stress that worsens DNA double-strand breaks in E. coli K1-infected neurons, leading to H3K4 trimethylation and phosphorylation of alpha-synuclein. Our findings suggest therapeutic strategies for post-bacterial meningitis treatment to potentially prevent the initiation of synucleinopathy.
Collapse
Affiliation(s)
- Van Thi Ai Tran
- Institute of Biophysics, Sungkyunkwan University, Suwon, South Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, South Korea
| | - Xiaohui Zhu
- Institute of Biophysics, Sungkyunkwan University, Suwon, South Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, South Korea
| | - Ariunzaya Jamsranjav
- Institute of Biophysics, Sungkyunkwan University, Suwon, South Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, South Korea
| | - Luke P Lee
- Institute of Biophysics, Sungkyunkwan University, Suwon, South Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, South Korea.
- Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Harvard Institute of Medicine, Harvard University, Boston, MA, USA.
| | - Hansang Cho
- Institute of Biophysics, Sungkyunkwan University, Suwon, South Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, South Korea.
- Department of Intelligent Healthcare Medicine, Sungkyunkwan University, Suwon, South Korea.
| |
Collapse
|
4
|
Steczkiewicz K, Kossakowski A, Janik S, Muszewska A. Low-complexity regions in fungi display functional groups and are depleted in positively charged amino acids. NAR Genom Bioinform 2025; 7:lqaf014. [PMID: 40041205 PMCID: PMC11878562 DOI: 10.1093/nargab/lqaf014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/29/2025] [Accepted: 02/20/2025] [Indexed: 03/06/2025] Open
Abstract
Reports on the diversity and occurrence of low-complexity regions (LCR) in Eukaryota are limited. Some studies have provided a more extensive characterization of LCR proteins in prokaryotes. There is a growing body of knowledge about a plethora of biological functions attributable to LCRs. However, it is hard to determine to what extent observed phenomena apply to fungi since most studies of fungal LCRs were limited to model yeasts. To fill this gap, we performed a survey of LCRs in proteins across all fungal tree of life branches. We show that the abundance of LCRs and the abundance of proteins with LCRs are positively correlated with proteome size. We observed that most LCRs are present in proteins with protein domains but do not overlap with the domain regions. LCRs are associated with many duplicated protein domains. The quantity of particular amino acids in LCRs deviates from the background frequency with a clear over-representation of amino acids with functional groups and a negative charge. Moreover, we discovered that each lineage of fungi favors distinct LCRs expansions. Early diverging fungal lineages differ in LCR abundance and composition pointing at a different evolutionary trajectory of each fungal group.
Collapse
Affiliation(s)
- Kamil Steczkiewicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5A, 02-106 Warsaw, Poland
| | - Aleksander Kossakowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5A, 02-106 Warsaw, Poland
| | - Stanisław Janik
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5A, 02-106 Warsaw, Poland
- Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Stefana Banacha 2, 02-097 Warsaw, Poland
| | - Anna Muszewska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5A, 02-106 Warsaw, Poland
| |
Collapse
|
5
|
Shen Y, Li Y, Quan Y, Jin W, Wang Y, Liu B, Wang Y. Effects of environment regulating T4SS on virulence and adaptability of Streptococcus suis. ENVIRONMENTAL RESEARCH 2025; 268:120751. [PMID: 39778619 DOI: 10.1016/j.envres.2025.120751] [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: 10/21/2024] [Revised: 12/30/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025]
Abstract
Streptococcus suis (S. suis) represents a significant bacterial pathogen, with its zoonotic transmission from infected or deceased pigs to humans posing a serious threat to public health. The type IV secretion system (T4SS), a critical virulence factor of S. suis, is tightly regulated by diverse environmental conditions. This study explores the influence of environmental variables, including temperature, incubation duration, monosaccharides, and metal ions, on the regulation of T4SS in S. suis and its associated pathogenicity. Results revealed that T4SS expression peaked during the stabilization phase at 37 °C, with galactose markedly enhancing T4SS expression relative to glucose. Zinc ions specifically enhanced the expression of the T4SS effector SspA among various metal ions. Moreover, zinc exposure significantly augmented both T4SS and virulence gene expression capabilities in S. suis. Zinc-treated S. suis exhibited enhanced adhesion, invasion, and colonization capacity in Hep-2 cells, Raw264.7 cells, and mouse models. These findings provide a deeper comprehension of the environmental modulation of T4SS in S. suis, paving the way for advanced studies into its mechanisms of pathogenicity.
Collapse
Affiliation(s)
- Yamin Shen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Engineering Research Center of Livestock and Poultry Emerging Disease Detection and Control, Luoyang, China
| | - Yue Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Engineering Research Center of Livestock and Poultry Emerging Disease Detection and Control, Luoyang, China
| | - Yingying Quan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Engineering Research Center of Livestock and Poultry Emerging Disease Detection and Control, Luoyang, China
| | - Wenjie Jin
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Engineering Research Center of Livestock and Poultry Emerging Disease Detection and Control, Luoyang, China
| | - Yuxin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Engineering Research Center of Livestock and Poultry Emerging Disease Detection and Control, Luoyang, China
| | - Baobao Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Engineering Research Center of Livestock and Poultry Emerging Disease Detection and Control, Luoyang, China.
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Engineering Research Center of Livestock and Poultry Emerging Disease Detection and Control, Luoyang, China.
| |
Collapse
|
6
|
Zhao T, Pellegrini L, van der Hee B, Boekhorst J, Fernandes A, Brugman S, van Baarlen P, Wells JM. Choroid plexus organoids reveal mechanisms of Streptococcus suis translocation at the blood-cerebrospinal fluid barrier. Fluids Barriers CNS 2025; 22:14. [PMID: 39930492 PMCID: PMC11812244 DOI: 10.1186/s12987-025-00627-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
Streptococcus suis is a globally emerging zoonotic pathogen that can cause invasive disease commonly associated with meningitis in pigs and humans. To cause meningitis, S. suis must invade the central nervous system (CNS) by crossing the neurovascular unit, also known as the blood-brain barrier (BBB), or vascularized choroid plexus (ChP) epithelium known as the blood-cerebrospinal fluid barrier (BCSFB). Recently developed ChP organoids have been shown to accurately replicate the cytoarchitecture and physiological functions of the ChP epithelium in vivo. Here, we used human induced pluripotent stem cells (iPSC)-derived ChP organoids as an in vitro model to investigate S. suis interaction and infection at the BCSFB. Our study revealed that S. suis is capable of translocating across the epithelium of ChP organoids without causing significant cell death or compromising the barrier integrity. Plasminogen (Plg) binding to S. suis in the presence of tissue plasminogen activator (tPA), which converts immobilized Plg to plasmin (Pln), significantly increased the basolateral to apical translocation across ChP organoids into the CSF-like fluid in the lumen. S. suis was able to replicate at the same rate in CSF and laboratory S. suis culture medium but reached a lower final density. The analysis of transcriptomes in ChP organoids after S. suis infection indicated inflammatory responses, while the addition of Plg further suggested extracellular matrix (ECM) remodeling. To our knowledge, this is the first study using ChP organoids to investigate bacterial infection of the BCSFB. Our findings highlight the potential of ChP organoids as a valuable tool for studying the mechanisms of bacterial interaction and infection of the human ChP in vitro.
Collapse
Affiliation(s)
- Tiantong Zhao
- Host-Microbe Interactomics, Department Animal Science, Wageningen University & Research, De Elst 1, Wageningen, 6708 WD, The Netherlands
| | - Laura Pellegrini
- Centre for Developmental Neurobiology, King's College London, Guys Campus, New Hunt's House, London, UK
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Bart van der Hee
- Host-Microbe Interactomics, Department Animal Science, Wageningen University & Research, De Elst 1, Wageningen, 6708 WD, The Netherlands
| | - Jos Boekhorst
- Host-Microbe Interactomics, Department Animal Science, Wageningen University & Research, De Elst 1, Wageningen, 6708 WD, The Netherlands
| | - Aline Fernandes
- Host-Microbe Interactomics, Department Animal Science, Wageningen University & Research, De Elst 1, Wageningen, 6708 WD, The Netherlands
| | - Sylvia Brugman
- Host-Microbe Interactomics, Department Animal Science, Wageningen University & Research, De Elst 1, Wageningen, 6708 WD, The Netherlands
| | - Peter van Baarlen
- Host-Microbe Interactomics, Department Animal Science, Wageningen University & Research, De Elst 1, Wageningen, 6708 WD, The Netherlands
| | - Jerry M Wells
- Host-Microbe Interactomics, Department Animal Science, Wageningen University & Research, De Elst 1, Wageningen, 6708 WD, The Netherlands.
| |
Collapse
|
7
|
Wu LL, Shi WD, Peng WF, Li GY. Unraveling the interplay between meningitis and mitochondria: Etiology, pathogenesis, and therapeutic insights. Int Immunopharmacol 2025; 147:113985. [PMID: 39765004 DOI: 10.1016/j.intimp.2024.113985] [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: 07/25/2024] [Revised: 12/11/2024] [Accepted: 12/28/2024] [Indexed: 01/29/2025]
Abstract
Meningitis, characterized by an inflammatory response affecting the membranes surrounding the brain and spinal cord, poses a formidable challenge to global public health. Its etiology spans a spectrum of infectious agents, ranging from bacteria, to viruses, fungi, and parasites. Concurrently, mitochondria-traditionally known as 'cellular powerhouses'-have emerged as critical players in various essential biological functions, including but not limited to, energy production, metabolic regulation, and cell fate determination. Emerging evidence suggests that mitochondria may play vital roles in the pathogenesis of meningitis. In this review, we delineated the definition, classification, etiology, pathogenesis, and clinical manifestations of meningitis, and elucidated the structure, dynamics and functions of mitochondria. We subsequently delved into the intricate interplay between meningitis and mitochondria, identifying potential therapeutic interventions targeting mitochondria for the first time. With clinical trials on the horizon, our review lays the foundation for a transformative era in meningitis therapeutics, where unraveling the intricate interplay between meningitis and mitochondria offers promise for mitigating neuroinflammation and improving patient outcomes.
Collapse
Affiliation(s)
- Li-Li Wu
- Department of Encephalopathy, Zhoukou Hospital of Traditional Chinese Medicine, Zhoukou 466099, China.
| | - Wei-Dong Shi
- Department of Orthopedics, Zhoukou Hospital of Traditional Chinese Medicine, Zhoukou 466099, China.
| | - Wei-Feng Peng
- Department of Encephalopathy, Zhoukou Hospital of Traditional Chinese Medicine, Zhoukou 466099, China; College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466000, China.
| | - Guo-Yin Li
- Department of Encephalopathy, Zhoukou Hospital of Traditional Chinese Medicine, Zhoukou 466099, China; College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466000, China; Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an 710062, China; Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
8
|
Alexander NG, Cutts WD, Hooven TA, Kim BJ. Mechanisms and Manifestations of Group B Streptococcus Meningitis in Newborns. J Pediatric Infect Dis Soc 2025; 14:piae103. [PMID: 39927629 PMCID: PMC11808573 DOI: 10.1093/jpids/piae103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/02/2024] [Indexed: 02/11/2025]
Abstract
Group B Streptococcus (GBS; Streptococcus agalactiae) is a gram-positive colonizer of the healthy intestinal and genitourinary microbiota. During and shortly after birth, neonates and infants can be opportunistically infected leading to sepsis, pneumonia, or meningitis among other illnesses. GBS is the leading cause of neonatal meningitis globally, and while prophylactic treatments have been successful for reducing early-onset disease, no decrease in the incidence of late-onset disease has occurred and no vaccine is currently available. In this review, we describe GBS both from a clinical and molecular standpoint. We first describe the history of GBS perinatal disease and its clinical presentation and treatment, as well as patient outcomes. We then present recently discovered GBS interactions at the blood-brain barrier that contribute to disease and inflammatory responses, and efforts to develop a broadly effective GBS vaccine.
Collapse
Affiliation(s)
- Natalie G Alexander
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - William D Cutts
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Thomas A Hooven
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Richard King Mellon Institute for Pediatric Research, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Brandon J Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa, Alabama, USA
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, Alabama, USA
| |
Collapse
|
9
|
Wang J, Kang L, Xu W, Xiao J, Min Y, Li S, Zhou C, Yin Y, Zhang X, Zhang Q. Progranulin Plays a Protective Role in Pneumococcal Meningitis by Inhibiting Pyroptosis. Immun Inflamm Dis 2025; 13:e70140. [PMID: 39887961 PMCID: PMC11783684 DOI: 10.1002/iid3.70140] [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: 06/22/2024] [Revised: 11/19/2024] [Accepted: 01/19/2025] [Indexed: 02/01/2025] Open
Abstract
OBJECTIVE Pneumococcal meningitis is a serious infectious disease with a high mortality rate and a global presence, and survivors have different degrees of neurological sequelae as a consequence of the host response to the infection. Progranulin (PGRN) is a multifunctional autocrine growth factor that is also a major immunoregulator. We want to investigate the role for PGRN in Pneumococcal meningitis in vivo and in vitro. METHOD Mouse and cell models were established to explore the protective effect and mechanism of PGRN against pneumococcal meningitis. RESULTS Progranulin plays a protective role in pneumococcal meningitis by inhibiting pyroptosis. Pyroptosis resulted from exposure of BV-2 cells to the bacterium and this was confirmed in the in vivo model. Administration of the NLRP3 inflammasome inhibitor MCC950 to mice prior to infection inhibited pyroptosis and protected PGRN -/- mice and BV-2 cell model from meningitis. CONCLUSION This study implicates a protective role for PGRN in pneumococcal meningitis by inhibiting pyroptosis, indicating that PGRN may have therapeutic potential.
Collapse
Affiliation(s)
- Jingyao Wang
- Department of Clinical 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, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory DiseasesChongqing Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory MedicineChongqing Medical UniversityChongqingChina
- Chengdu Women's and Children's Central Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Lihua Kang
- Department of Clinical 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, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory DiseasesChongqing Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory MedicineChongqing Medical UniversityChongqingChina
- Department of Clinical LaboratoryWomen and Children's Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Wenlong Xu
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Jiangming Xiao
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Yajun Min
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Sijie Li
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Changlong Zhou
- Department of Neurosurgery, Yongchuan HospitalChongqing Medical UniversityChongqingPeople's Republic of China
| | - Yibing Yin
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Xuemei Zhang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Department of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Qun Zhang
- Department of Clinical 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, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory DiseasesChongqing Medical UniversityChongqingPeople's Republic of China
| |
Collapse
|
10
|
Ma J, Wu H, Ma Z, Wu Z. Bacterial and host factors involved in zoonotic Streptococcal meningitis. Microbes Infect 2025; 27:105335. [PMID: 38582147 DOI: 10.1016/j.micinf.2024.105335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024]
Abstract
Zoonotic streptococci cause several invasive diseases with high mortality rates, especially meningitis. Numerous studies elucidated the meningitis pathogenesis of zoonotic streptococci, some specific to certain bacterial species. In contrast, others are shared among different bacterial species, involving colonization and invasion of mucosal barriers, survival in the bloodstream, breaching the blood-brain and/or blood-cerebrospinal fluid barrier to access the central nervous system, and triggering inflammation of the meninges. This review focuses on the recent advancements in comprehending the molecular and cellular events of five major zoonotic streptococci responsible for causing meningitis in humans or animals, including Streptococcus agalactiae, Streptococcus equi subspecies zooepidemicus, Streptococcus suis, Streptococcus dysgalactiae, and Streptococcus iniae. The underlying mechanism was summarized into four themes, including 1) bacterial survival in blood, 2) brain microvascular endothelial cell adhesion and invasion, 3) penetration of the blood-brain barrier, and 4) activation of the immune system and inflammatory reaction within the brain. This review may contribute to developing therapeutics to prevent or mitigate injury of streptococcal meningitis and improve risk stratification.
Collapse
Affiliation(s)
- Jiale Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China; WOAH Reference Lab for Swine Streptococcosis, Nanjing 210014, China
| | - Huizhen Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China; WOAH Reference Lab for Swine Streptococcosis, Nanjing 210014, China
| | - Zhe Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Zongfu Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210014, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210014, China; WOAH Reference Lab for Swine Streptococcosis, Nanjing 210014, China; Guangdong Provincial Key Laboratory of Research on the Technology of Pig-breeding and Pig-disease Prevention, Guangzhou 511400, China.
| |
Collapse
|
11
|
Vollmuth N, Sauerwein T, Foerstner KU, Westermann AJ, Schubert-Unkmeir A, Lam DW, Kim BJ. Streptococcus agalactiae strain COH1 transcriptome in association with stem cell-derived brain-like endothelial cells. Microbiol Resour Announc 2024; 13:e0045524. [PMID: 39526785 PMCID: PMC11636360 DOI: 10.1128/mra.00455-24] [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: 06/25/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024] Open
Abstract
Streptococcus agalactiae (Group B Streptococcus) strain COH1 is a representative strain of serotype III, multi-locus sequence type 17, which is disproportionately associated with neonatal meningitis. Here we report the transcriptome of COH1 when interacting with human brain endothelial cells compared with COH1 alone.
Collapse
Affiliation(s)
- Nadine Vollmuth
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Till Sauerwein
- Institute of Molecular Infection Biology (IMIB), University of Wuerzburg, Wuerzburg, Germany
- ZB MED, Information Centre for Life Sciences, Cologne, Germany
| | - Konrad U. Foerstner
- Institute of Molecular Infection Biology (IMIB), University of Wuerzburg, Wuerzburg, Germany
- ZB MED, Information Centre for Life Sciences, Cologne, Germany
- TH Koeln, University of Applied Sciences, Cologne, Germany
| | - Alexander J. Westermann
- Institute of Molecular Infection Biology (IMIB), University of Wuerzburg, Wuerzburg, Germany
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Wuerzburg, Germany
| | | | - Daryl W. Lam
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Brandon J. Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
- Department of Microbiology, University of Alabama at Birmingham Heesink School of Medicine, Birmingham, Alabama, USA
- University of Alabama Center of Convergent Biosciences and Medicine, Tuscaloosa, Alabama, USA
- University of Alabama Life Research, Tuscaloosa, Alabama, USA
| |
Collapse
|
12
|
Al-Obaidi MMJ, Al Siyabi MSK, Muthanna A, Mohd Desa MN. Understanding the mechanisms of Streptococcus pneumoniae in penetrating the blood-brain barrier: insights into bacterial binding with central nervous system host receptors. Tissue Barriers 2024:2434764. [PMID: 39629682 DOI: 10.1080/21688370.2024.2434764] [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: 07/14/2024] [Revised: 11/12/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024] Open
Abstract
This review investigates the pathogenic processes through which Streptococcus pneumoniae crosses the blood-brain barrier (BBB) to cause meningitis, with a focus on the interaction with host receptors in the central nervous system (CNS). S. pneumoniae a primary cause of bacterial meningitis, utilizes unique receptor-mediated pathways to infiltrate the BBB. The bacterial interaction with the platelet-activating factor receptor (PAFR) and the polymeric immunoglobulin receptor (pIgR) is looked at in this study. The goal is to understand how this interaction helps the bacterium move across the BBB and cause infection in the CNS. We examine the functions of cellular and molecular participants at the endothelium level, such as cytokines, chemokines, and matrix metalloproteinases (MMP), which have a role in the development of the disease. This study consolidates data from multiple studies, providing a thorough summary of the interactions between S. pneumoniae and the BBB. It also explores potential treatment targets that could reduce the significant illness and death rates associated with pneumococcal meningitis.
Collapse
Affiliation(s)
- Mazen M Jamil Al-Obaidi
- University of Technology and Applied Sciences, Rustaq, College of Education, Science Department, Sultanate of Oman
| | - Muzna Saif Khalfan Al Siyabi
- University of Technology and Applied Sciences, College of applied sciences and pharmacy, Department of applied sciences, Biology section, Muscat, Sultanate of Oman
| | - AbdulRahman Muthanna
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Mohd Nasir Mohd Desa
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
13
|
Xu Y, Wang J, Qin X, Liu J. Advances in the pathogenesis and treatment of pneumococcal meningitis. Virulence 2024; 15:2387180. [PMID: 39192572 PMCID: PMC11364070 DOI: 10.1080/21505594.2024.2387180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/04/2024] [Accepted: 07/28/2024] [Indexed: 08/29/2024] Open
Abstract
Streptococcus pneumoniae is a common pathogen associated with community-acquired bacterial meningitis, characterized by high morbidity and mortality rates. While vaccination reduces the incidence of meningitis, many survivors experience severe brain damage and corresponding sequelae. The pathogenesis of pneumococcal meningitis has not been fully elucidated. Currently, meningitis requires bacterial disruption of the blood - brain barrier, a process that involves the interaction of bacterial surface components with host cells and various inflammatory responses. This review delineates the global prevalence, pathogenesis, and treatment strategies of pneumococcal meningitis. The objective is to enhance the thorough comprehension of the clinical manifestations and biological mechanisms of the disease, thereby enabling more efficient prevention, diagnosis, and therapeutic interventions.
Collapse
Affiliation(s)
- Yiyun Xu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Ji Wang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Xiaosong Qin
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Jianhua Liu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| |
Collapse
|
14
|
Deng S, Liao J, Li H, Xu J, Fan J, Xia J, Wang J, Lei L, Chen M, Han Y, Zhai R, Zhou C, Zhou R, Cheng C, Song H. Streptococcus suis subtilisin-like serine proteases SspA-1 and SspA-2 interplay with complement C3a and C5a to facilitate bacterial immune evasion and infection. Virulence 2024; 15:2301246. [PMID: 38170683 PMCID: PMC10795781 DOI: 10.1080/21505594.2023.2301246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/28/2023] [Indexed: 01/05/2024] Open
Abstract
Streptococcus suis (S. suis), a significant zoonotic bacterial pathogen impacting swine and human, is associated with severe systemic diseases such as streptococcal toxic shock-like syndrome, meningitis, septicaemia, and abrupt fatality. The multifaceted roles of complement components C5a and C3a extend to orchestrating inflammatory cells recruitment, oxidative burst induction, and cytokines release. Despite the pivotal role of subtilisin-like serine proteases in S. suis pathogenicity, their involvement in immune evasion remains underexplored. In the present study, we identify two cell wall-anchored subtilisin-like serine proteases in S. suis, SspA-1 and SspA-2, as binding partners for C3a and C5a. Through Co-Immunoprecipitation, Enzyme-Linked Immunosorbent and Far-Western Blotting Assays, we validate their interactions with the aforementioned components. However, SspA-1 and SspA-2 have no cleavage activity against complement C3a and C5a performed by Cleavage assay. Chemotaxis assays reveal that recombinant SspA-1 and SspA-2 effectively attenuate monocyte chemotaxis towards C3a and C5a. Notably, the ΔsspA-1, ΔsspA-1, and ΔsspA-1/2 mutant strains exhibit compromised survival in blood, and resistance of opsonophagocytosis, alongside impaired survival in blood and in vivo colonization compared to the parental strain SC-19. Critical insights from the murine and Galleria mellonella larva infection models further underscore the significance of sspA-1 in altering mortality rates. Collectively, our findings indicate that SspA-1 and SspA-2 are novel binding proteins for C3a and C5a, thereby shedding light on their pivotal roles in S. suis immune evasion and the pathogenesis.
Collapse
Affiliation(s)
- Simin Deng
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, P.R. China
| | - Junhui Liao
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, P.R. China
| | - Haojie Li
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, P.R. China
| | - Jiali Xu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, P.R. China
| | - Jingyan Fan
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, P.R. China
| | - Jing Xia
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, P.R. China
| | - Jing Wang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, P.R. China
| | - Lei Lei
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, P.R. China
| | - Mianmian Chen
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, P.R. China
| | - Yue Han
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, P.R. China
| | - Ruidong Zhai
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, P.R. China
| | - Chang Zhou
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, P.R. China
| | - Rui Zhou
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Changyong Cheng
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, P.R. China
| | - Houhui Song
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection & Internet Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang Province, P.R. China
| |
Collapse
|
15
|
Maghembe RS, Magulye MA, Makaranga A, Nsubuga G, Sekyanzi S, Moto EA, Mwesigwa S, Katagirya E. Metagenome mining divulges virulent and multidrug resistant Pseudomonas aeruginosa ST242 and Klebsiella michiganensis ST∗1b23 coinfecting an 8-month-old meningitis infant under ICU in Kampala, Uganda, East Africa. Heliyon 2024; 10:e39455. [PMID: 39498086 PMCID: PMC11532849 DOI: 10.1016/j.heliyon.2024.e39455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 11/07/2024] Open
Abstract
Pediatric meningitis is a global health problem, with insufficiently known pathogens and antibiotic resistance (AMR) especially in low-resource settings. Here, we sought to uncover the virulence and AMR of pathogens associated with infant meningitis, treated with ceftriaxone, in Kampala, Uganda. In a bid to isolate Klebsiella oxytoca, we coincidentally recovered a co-culture and challenged it with antibiotic susceptibility testing (AST) on a panel of 14 antibiotics. We then combined metagenome binning with antiSMASH/PRISM genome mining to unveil the pathogens, their virulence and molecular targets in relation to meningitis. From AST, the co-culture exhibited resistance to multiple aminoglycosides, fluroquinolones, and β-lactams, including ceftriaxone, the inherently used drug. From metagenome annotation, the first bin was identified as Pseudomonas aeruginosa ST242 and the second as Klebsiella michiganensis ST∗1b23. Among others, P. aeruginosa ST242 virulence factors include type 3 and type 6 secretion systems, biofilm, and nonribosomal peptides (NRPs) of the pyoverdine synthase operon, targeting claudin-5, a component of the tight junctions of the blood-brain barrier (BBB). The P. aeruginosa ST242 genome portrays intrinsic resistance to beta-lactamases (blaOXA-50 and blaPAO), aminoglycosides [aph(3')-IIb)], fluoroquinolones (crpP), tetracycline (tmexD2) and fosfomycin (fosA), among others. From K. michiganensis ST∗1b23 genome mining we elucidated a yersiniabactin-related metabolite, targeting the ligand-binding domain of the human polymeric immunoglobulin receptor (pIgR) and other components of the BBB. The K. michiganensis ST∗1b23 chromosome encodes the genes blaOXY-1 and OqxA/B, conferring resistance to β-lactams, fluoroquinolones, and trimethoprim respectively. Notably, we found one frameshift and nine substitution mutations conferring carbapenem and cephalosporin resistance mechanisms. Overall, our findings strongly suggest coinfection and a mechanistic crosstalk between P. aeruginosa ST242 and K. michiganensis ST∗1b23 in the pathogenesis of meningitis in this case. Importantly, ceftriaxone could be an inappropriate treatment choice for these pathogens. Hence, serious surveillance and experimental studies will improve the management of pediatric meningitis.
Collapse
Affiliation(s)
- Reuben S. Maghembe
- Department of Microbiology and Parasitology, Faculty of Medicine, St. Francis University College of Health and Allied Sciences (SFUCHAS), P. O. Box 175, Ifakara, Tanzania
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
- Department of Microbiology and Immunology, Faculty of Biomedical Sciences, Kampala International University-Western Campus (KIU-WC), Ishaka, Bushenyi, Uganda
- Department of Health and Biomedical Sciences, Didia Education and Health Organization (DEHO), P. O. Box 113, Shinyanga, Tanzania
| | - Maximilian A.K. Magulye
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
- Department of Health and Biomedical Sciences, Didia Education and Health Organization (DEHO), P. O. Box 113, Shinyanga, Tanzania
- Department of Medical Microbiology, College of Health Sciences, Makerere University, P. O. Box 7072, Kampala, Uganda
| | - Abdalah Makaranga
- Department of Health and Biomedical Sciences, Didia Education and Health Organization (DEHO), P. O. Box 113, Shinyanga, Tanzania
| | - Gideon Nsubuga
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Simon Sekyanzi
- Department of Medical Microbiology, College of Health Sciences, Makerere University, P. O. Box 7072, Kampala, Uganda
| | - Edward A. Moto
- Department of Biology, College of Natural and Mathematical Sciences, University of Dodoma, Dodoma, Tanzania
| | - Savannah Mwesigwa
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Eric Katagirya
- Department of Immunology and Molecular Biology, College of Health Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
16
|
Zheng Y, Sun H, Wang Y, Jin C, Li X, Pang Y, Ge Q, Wang L, Liu B. CsiR-Mediated Signal Transduction Pathway in Response to Low Iron Conditions Promotes Escherichia coli K1 Invasion and Penetration of the Blood-Brain Barrier. J Infect Dis 2024; 230:e807-e817. [PMID: 38531686 PMCID: PMC11481304 DOI: 10.1093/infdis/jiae157] [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: 07/28/2023] [Revised: 03/16/2024] [Accepted: 03/26/2024] [Indexed: 03/28/2024] Open
Abstract
Escherichia coli K1 is the leading cause of neonatal gram-negative bacterial meningitis, but the pathogenesis of E coli K1 meningitis remains unclear. Blood-brain barrier (BBB) penetration is a crucial step in E coli meningitis development. Here, we uncovered the crucial role of CsiR, a GntR family regulator, in E coli K1 virulence. During infection, csiR expression was induced due to the derepression by Fur in the blood and human brain microvascular endothelial cells (HBMECs). CsiR positively regulated ilvB expression, which is associated with branched chain amino acid synthesis. Furthermore, we revealed that IlvB activated the FAK/PI3K pathway of HBMECs to induce actin cytoskeleton rearrangements, thereby promoting the bacterial invasion and penetration of the BBB. Overall, this study reveals a CsiR-mediated virulence regulation pathway in E coli K1, which may provide a useful target for the prevention or therapy of E coli meningitis.
Collapse
Affiliation(s)
- Yangyang Zheng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University,Tianjin, People's Republic of China
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, People's Republic of China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - Hao Sun
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University,Tianjin, People's Republic of China
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, People's Republic of China
| | - Yanling Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University,Tianjin, People's Republic of China
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, People's Republic of China
| | - Chen Jin
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University,Tianjin, People's Republic of China
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, People's Republic of China
| | - Xiaoya Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University,Tianjin, People's Republic of China
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, People's Republic of China
| | - Yu Pang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University,Tianjin, People's Republic of China
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, People's Republic of China
| | - Qianwen Ge
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University,Tianjin, People's Republic of China
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, People's Republic of China
| | - Lei Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University,Tianjin, People's Republic of China
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, People's Republic of China
| | - Bin Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University,Tianjin, People's Republic of China
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Tianjin, People's Republic of China
- Nankai International Advanced Research Institute, Shenzhen, People's Republic of China
| |
Collapse
|
17
|
Ibrahim A, Saleem N, Naseer F, Ahmed S, Munawar N, Nawaz R. From cytokines to chemokines: Understanding inflammatory signaling in bacterial meningitis. Mol Immunol 2024; 173:117-126. [PMID: 39116800 DOI: 10.1016/j.molimm.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 06/11/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024]
Abstract
Bacterial meningitis is a serious central nervous system (CNS) infection, claiming millions of human lives annually around the globe. The deadly infection involves severe inflammation of the protective sheath of the brain, i.e., meninges, and sometimes also consists of the brain tissue, called meningoencephalitis. Several inflammatory pathways involved in the pathogenesis of meningitis caused by Streptococcus pneumoniae, Neisseria meningitidis, Escherichia coli, Haemophilus influenzae, Mycobacterium tuberculosis, Streptococcus suis, etc. are mentioned in the scientific literature. Many in-vitro and in-vivo analyses have shown that after the disruption of the blood-brain barrier (BBB), these pathogens trigger several inflammatory pathways including Toll-Like Receptor (TLR) signaling in response to Pathogen-Associated Molecular Patterns (PAMPs), Nucleotide oligomerization domain (NOD)-like receptor-mediated signaling, pneumolysin related signaling, NF-κB signaling and many other pathways that lead to pro-inflammatory cascade and subsequent cytokine release including interleukine (IL)-1β, tumor necrosis factor(TNF)-α, IL-6, IL-8, chemokine (C-X-C motif) ligand 1 (CXCL1) along with other mediators, leading to neuroinflammation. The activation of another protein complex, nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome, also takes place resulting in the maturation and release of IL-1β and IL-18, hence potentiating neuroinflammation. This review aims to outline the inflammatory signaling pathways associated with the pathogenesis of bacterial meningitis leading to extensive pathological changes in neurons, astrocytes, oligodendrocytes, and other central nervous system cells.
Collapse
Affiliation(s)
- Ahsan Ibrahim
- Shifa College of Pharmaceutical Sciences, Shifa Tameer e Millat University, Islamabad, Pakistan
| | - Nida Saleem
- Shifa College of Pharmaceutical Sciences, Shifa Tameer e Millat University, Islamabad, Pakistan
| | - Faiza Naseer
- Shifa College of Pharmaceutical Sciences, Shifa Tameer e Millat University, Islamabad, Pakistan; Department of Biosciences, Shifa Tameer e Millat University, Islamabad, Pakistan.
| | - Sagheer Ahmed
- Shifa College of Pharmaceutical Sciences, Shifa Tameer e Millat University, Islamabad, Pakistan.
| | - Nayla Munawar
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rukhsana Nawaz
- Department of Clinical Psychology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
18
|
Fernandes DC, Eto SF, Baldassi AC, Balbuena TS, Charlie-Silva I, de Andrade Belo MA, Pizauro JM. Meningitis caused by Aeromonas hydrophila in Oreochromis niloticus: Proteomics and druggability of virulence factors. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109687. [PMID: 38866348 DOI: 10.1016/j.fsi.2024.109687] [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: 03/18/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024]
Abstract
Meningitis caused by Gram-negative bacteria is a serious public health problem, causing morbidity and mortality in both children and adults. Here, we propose a novel experimental model using Nile tilapia (Oreochromis niloticus) to study neuroinflammation. The fish were infected with Aeromonas hydrophila, and the course of infection was monitored in the peripheral blood. Septicemia was obvious in the blood, while in the brain tissue, infection of the meninges was present. The histopathological examination showed suppurative meningitis, and the cellular immune response in the brain tissue during infection was mediated by microglia. These cells were morphologically characterized and phenotyped by MHC class II markers and CD68. The increased production of TNF-α, IL-1β and iNOS supported the infiltration of these cells during the neuroinflammatory process. In the proteomic analysis of A. hydrophila isolated from brain tissue, we found chemotactic and transport proteins, proteolytic enzymes and enzymes associated with the dismutation of nitric oxide (NO), as well as motor proteins and those responsible for cell division. After characterizing the most abundant proteins during the course of infection, we investigated the druggability index of these proteins and identified promising peptide sequences as molecular targets that are similar among bacteria. Thus, these findings deepened the understanding of the pathophysiology of meningitis caused by A. hydrophila. Moreover, through the proteomics analysis, important mechanisms and pathways used by the pathogen to subvert the host response were revealed, providing insights for the development of novel antibiotics and vaccines.
Collapse
Affiliation(s)
- Dayanne Carla Fernandes
- Institute of Chemistry, São Paulo State University (Unesp), Araraquara, Sao Paulo, SP, Brazil.
| | - Silas Fernandes Eto
- Laboratory Center of Excellence in New Target Discovery (CENTD) Special Laboratory, Butantan Institute, São Paulo, SP, Brazil
| | - Amanda Cristina Baldassi
- Department of Technology, School of Agrarian and Veterinary Sciences, São Paulo State University (Unesp), Jaboticabal, Sao Paulo, SP, Brazil
| | - Thiago Santana Balbuena
- Department of Technology, School of Agrarian and Veterinary Sciences, São Paulo State University (Unesp), Jaboticabal, Sao Paulo, SP, Brazil
| | - Ives Charlie-Silva
- Institute of Chemistry, São Paulo State University (Unesp), Araraquara, Sao Paulo, SP, Brazil
| | | | - João Martins Pizauro
- Department of Technology, School of Agrarian and Veterinary Sciences, São Paulo State University (Unesp), Jaboticabal, Sao Paulo, SP, Brazil
| |
Collapse
|
19
|
Satsantitham K, Sritangos P, Wet-osot S, Chudapongse N, Weeranantanapan O. Cellular uptake of allicin in the hCMEC/D3 human brain endothelial cells: exploring blood-brain barrier penetration in an in vitro model. PeerJ 2024; 12:e17742. [PMID: 39035169 PMCID: PMC11260074 DOI: 10.7717/peerj.17742] [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: 03/12/2024] [Accepted: 06/24/2024] [Indexed: 07/23/2024] Open
Abstract
Background Allicin, a bioactive compound derived from garlic (Allium sativum), demonstrates antibacterial activity against a broad spectrum of bacteria including the most common meningitis pathogens. In order to advocate for allicin as a potential therapeutic candidate for bacterial meningitis, the present study aimed to assess the ability of allicin to cross the blood-brain barrier (BBB) using an in vitro model. Methods The cell viability of the human brain endothelial cell line hCMEC/D3 after incubation with various concentrations of allicin was investigated using an MTT assay at 3 and 24 h. Additionally, reactive oxygen species (ROS) production of allicin-treated hCMEC/D3 cells was examined at 3 h. The concentrations of allicin that were not toxic to the cells, as determined by the MTT assay, and did not significantly increase ROS generation, were then used to investigate allicin's ability to traverse the in vitro BBB model for 3 h. High-performance liquid chromatography (HPLC) analysis was utilized to examine the allicin concentration capable of passing the in vitro BBB model. The cellular uptake experiments were subsequently performed to observe the uptake of allicin into hCMEC/D3 cells. The pkCSM online tool was used to predict the absorption, distribution, metabolism, excretion, and pharmacokinetic properties of allicin and S-allylmercaptoglutathione (GSSA). Results The results from MTT assay indicated that the highest non-toxicity concentration of allicin on hCMEC/D3 cells was 5 µg/ml at 3 h and 2 µg/ml at 24 h. Allicin significantly enhanced ROS production of hCMEC/D3 cells at 10 µg/ml at 3 h. After applying the non-toxicity concentrations of allicin (0.5-5 µg/ml) to the in vitro BBB model for 3 h, allicin was not detectable in both apical and basolateral chambers in the presence of hCMEC/D3 cells. On the contrary, allicin was detected in both chambers in the absence of the cells. The results from cellular uptake experiments at 3 h revealed that hCMEC/D3 cells at 1 × 104 cells could uptake allicin at concentrations of 0.5, 1, and 2 µg/ml. Moreover, allicin uptake of hCMEC/D3 cells was proportional to the cell number, and the cells at 5 × 104 could completely uptake allicin at a concentration of 5 µg/ml within 0.5 h. The topological polar surface area (TPSA) predicting for allicin was determined to be 62.082 Å2, indicating its potential ability to cross the BBB. Additionally, the calculated logBB value surpassing 0.3 suggests that the compound may exhibit ease of penetration through the BBB. Conclusion The present results suggested that allicin was rapidly taken up by hCMEC/D3 cells in vitro BBB model. The prediction results of allicin's distribution patterns suggested that the compound possesses the capability to enter the brain.
Collapse
Affiliation(s)
- Kankawi Satsantitham
- School of Preclinical Sciences, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Pishyaporn Sritangos
- School of Preclinical Sciences, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Sirawit Wet-osot
- Department of Medical Sciences, Ministry of Public Health, Nonthaburi, Thailand
| | | | - Oratai Weeranantanapan
- School of Preclinical Sciences, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| |
Collapse
|
20
|
Yaghoobi A, Abiri R, Alvandi A, Manouchehri I, Arkan E, Jalalvand AR. A novel biosensing strategy for identification of three important bacteria causing meningitis. J Microbiol Methods 2024; 222:106954. [PMID: 38754480 DOI: 10.1016/j.mimet.2024.106954] [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: 03/25/2024] [Revised: 05/11/2024] [Accepted: 05/12/2024] [Indexed: 05/18/2024]
Abstract
Bacterial meningitis is an acute infection which requires rapid diagnosis and treatment due to the high mortality and serious consequences of the disease. The purpose of this study was to design a homemade multiplex PCR and a novel fluorescence biosensor on chip (FBC) to detect three important agents of meningitis including Streptococcus pneumoniae (S. pneumoniae), Neisseria meningitidis (N. meningitidis), and Haemophilus influenzae (H. influenzae). The homemade multiplex PCR can diagnose three bacterial species simultaneously. Fabrication of FBC was carried out based on the deposition of lead nanoparticles on a quartz slide using the thermal evaporation method. Then, the SH-Cap Probe/Target ssDNA /FAM-Rep probe was loaded on lead film. The evaluation of the fluorescence reaction when the probes bind to the target ssDNA was assessed by a Cytation 5 Cell Imaging Multimode Reader Bio-Tek. The limit of detections (LOD) in homemade PCR and FBC to identify S. pneumoniae were 119 × 102 CFU/mL (0.27 ng/μL) and 380 CFU/mL (9 pg/μL), respectively. The LODs of homemade PCR and FBC for detection of N. meningitidis were 4.49 CFU/mL (1.1 pg/μL) and 13 × 103 CFU/mL (30 pg/μL), respectively. Our results confirmed the LODs of homemade PCR and FBC in detection of H. influenzae were 15.1 CFU/mL (30 fg/μL) and 41 × 102 CFU/mL (90 pg/ μL), respectively. Both techniques had appropriate sensitivity and specificity in detection of S. pneumoniae, N. meningitidis and H. influenzae.
Collapse
Affiliation(s)
- Azam Yaghoobi
- Department of Microbiology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Abiri
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Amirhoushang Alvandi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Iraj Manouchehri
- Department of Physic, Faculty of Sciences, Kurdistan University, Sanandaj, Iran
| | - Elham Arkan
- Nano Drug Delivery Research Center, Research Institute for Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali R Jalalvand
- Pharmaceutical Sciences Research Center, Health institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
21
|
Vollmuth N, Bridgers BE, Armstrong ML, Wood JF, Gildea AR, Espinal ER, Hooven TA, Barbieri G, Westermann AJ, Sauerwein T, Foerstner KU, Schubert-Unkmeir A, Kim BJ. Group B Streptococcus transcriptome when interacting with brain endothelial cells. J Bacteriol 2024; 206:e0008724. [PMID: 38771039 PMCID: PMC11332166 DOI: 10.1128/jb.00087-24] [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: 03/01/2024] [Accepted: 04/22/2024] [Indexed: 05/22/2024] Open
Abstract
Bacterial meningitis is a life-threatening infection of the central nervous system (CNS) that occurs when bacteria are able to cross the blood-brain barrier (BBB) or the meningeal-cerebrospinal fluid barrier (mBCSFB). The BBB and mBCSFB comprise highly specialized brain endothelial cells (BECs) that typically restrict pathogen entry. Group B Streptococcus (GBS or Streptococcus agalactiae) is the leading cause of neonatal meningitis. Until recently, identification of GBS virulence factors has relied on genetic screening approaches. Instead, we here conducted RNA-seq analysis on GBS when interacting with induced pluripotent stem cell-derived BECs (iBECs) to pinpoint virulence-associated genes. Of the 2,068 annotated protein-coding genes of GBS, 430 transcripts displayed significant changes in expression after interacting with BECs. Notably, we found that the majority of differentially expressed GBS transcripts were downregulated (360 genes) during infection of iBECs. Interestingly, codY, encoding a pleiotropic transcriptional repressor in low-G + C Gram-positive bacteria, was identified as being highly downregulated. We conducted qPCR to confirm the codY downregulation observed via RNA-seq during the GBS-iBEC interaction and obtained codY mutants in three different GBS background parental strains. As anticipated from the RNA-seq results, the [Formula: see text]codY strains were more adherent and invasive in two in vitro BEC models. Together, this demonstrates the utility of RNA-seq during the BEC interaction to identify GBS virulence modulators. IMPORTANCE Group B Streptococcus (GBS) meningitis remains the leading cause of neonatal meningitis. Research work has identified surface factors and two-component systems that contribute to GBS disruption of the blood-brain barrier (BBB). These discoveries often relied on genetic screening approaches. Here, we provide transcriptomic data describing how GBS changes its transcriptome when interacting with brain endothelial cells. Additionally, we have phenotypically validated these data by obtaining mutants of a select regulator that is highly down-regulated during infection and testing on our BBB model. This work provides the research field with a validated data set that can provide an insight into potential pathways that GBS requires to interact with the BBB and open the door to new discoveries.
Collapse
Affiliation(s)
- Nadine Vollmuth
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Bailey E. Bridgers
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Madelyn L. Armstrong
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Jacob F. Wood
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Abigail R. Gildea
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Eric R. Espinal
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Thomas A. Hooven
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Richard King Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Giulia Barbieri
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Alexander J. Westermann
- Institute of Molecular Infection Biology (IMIB), University of Wuerzburg, Wuerzburg, Germany
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Wuerzburg, Germany
| | - Till Sauerwein
- Institute of Molecular Infection Biology (IMIB), University of Wuerzburg, Wuerzburg, Germany
- ZB MED, Information Centre for Life Sciences, Cologne, Germany
| | - Konrad U. Foerstner
- Institute of Molecular Infection Biology (IMIB), University of Wuerzburg, Wuerzburg, Germany
- ZB MED, Information Centre for Life Sciences, Cologne, Germany
- TH Koeln, University of Applied Sciences, Cologne, Germany
| | | | - Brandon J. Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
- Department of Microbiology, University of Alabama at Birmingham Heesink School of Medicine, Birmingham, Alabama, USA
- University of Alabama Center of Convergent Biosciences and Medicine, Tuscaloosa, Alabama, USA
- University of Alabama Life Research, Tuscaloosa, Alabama, USA
| |
Collapse
|
22
|
Chekrouni N, van Soest TM, da Cruz Campos AC, Brouwer MC, van de Beek D. Bacterial load in cerebrospinal fluid predicts unfavourable outcome in pneumococcal meningitis: a prospective nationwide cohort study. Clin Microbiol Infect 2024; 30:772-778. [PMID: 38479702 DOI: 10.1016/j.cmi.2024.03.012] [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: 01/02/2024] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 04/01/2024]
Abstract
OBJECTIVES The objective of this study was to determine the role of cerebrospinal fluid (CSF) bacterial load in adults with pneumococcal meningitis. METHODS We quantified bacterial load in CSF samples from the diagnostic lumbar puncture of adults with community-acquired pneumococcal meningitis. We also measured CSF concentrations of complement component 5a (C5a), and determined associations between bacterial load, clinical characteristics, C5a and unfavourable outcome (Glasgow Outcome Scale score <5). RESULTS Bacterial load was quantified in 152 CSF samples. Median age of these patients was 61 years (interquartile range [IQR] 51-68), and 69 of 152 (45%) were female. Median CSF bacterial load was 1.6 × 104 DNA copies/mL (IQR 3.4 × 103-1.2 × 105), and did not correlate with CSF white cell count nor with CSF protein concentrations. Median CSF C5a concentration was 35.8 mg/L (IQR 15.9-105.6), and was moderately correlated with CSF bacterial loads (Spearman's rho = 0.42; p < 0001). High bacterial loads were associated with development of complications, such as circulatory shock (OR per logarithmic increase: 2.4, 95% CI: 2.0-2.9; p < 0.001) and cerebrovascular complications [OR: 1.9, 95% CI: 1.6-2.3; p < 0.001]). High bacterial loads were also associated with unfavourable outcome (OR: 2.8, 95% CI: 2.4-3.3; p < 0.001) and death (OR: 3.1, 95% CI: 2.6-3.8; p < 0.001). In a multivariable regression model including age, immunocompromised state, extrameningeal infection focus, admission Glasgow Coma Scale score and CSF C5a concentration, CSF bacterial load remained an independent predictor of unfavourable outcome (adjusted OR: 2.5, 95% CI: 1.6-3.9; p < 0.001). DISCUSSION High CSF bacterial load predicts the development of complications and unfavourable outcome in adults with pneumococcal meningitis.
Collapse
Affiliation(s)
- Nora Chekrouni
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef, Amsterdam, The Netherlands
| | - Thijs M van Soest
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef, Amsterdam, The Netherlands
| | - Ana C da Cruz Campos
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef, Amsterdam, The Netherlands
| | - Matthijs C Brouwer
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef, Amsterdam, The Netherlands
| | - Diederik van de Beek
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef, Amsterdam, The Netherlands.
| |
Collapse
|
23
|
Sereme Y, Schrimp C, Faury H, Agapoff M, Lefebvre-Wloszczowski E, Chang Marchand Y, Ageron-Ardila E, Panafieu E, Blec F, Coureuil M, Frapy E, Tsatsaris V, Bonacorsi S, Skurnik D. A live attenuated vaccine to prevent severe neonatal Escherichia coli K1 infections. Nat Commun 2024; 15:3021. [PMID: 38589401 PMCID: PMC11001983 DOI: 10.1038/s41467-024-46775-x] [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: 08/25/2023] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
Preterm birth is currently the leading cause of neonatal morbidity and mortality. Genetic, immunological and infectious causes are suspected. Preterm infants have a higher risk of severe bacterial neonatal infections, most of which are caused by Escherichia coli an in particular E. coli K1strains. Women with history of preterm delivery have a high risk of recurrence and therefore constitute a target population for the development of vaccine against E. coli neonatal infections. Here, we characterize the immunological, microbiological and protective properties of a live attenuated vaccine candidate in adult female mice and their pups against after a challenge by K1 and non-K1 strains of E. coli. Our results show that the E. coli K1 E11 ∆aroA vaccine induces strong immunity, driven by polyclonal bactericidal antibodies. In our model of meningitis, mothers immunized prior to mating transfer maternal antibodies to pups, which protect newborn mice against various K1 and non-K1 strains of E. coli. Given the very high mortality rate and the neurological sequalae associated with neonatal E. coli K1 meningitis, our results constitute preclinical proof of concept for the development of a live attenuated vaccine against severe E. coli infections in women at risk of preterm delivery.
Collapse
Affiliation(s)
- Youssouf Sereme
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades, Paris, France
| | - Cécile Schrimp
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades, Paris, France
| | - Helène Faury
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades, Paris, France
- Department of Microbiology, Necker Hospital, University de Paris, Paris, France
| | - Maeva Agapoff
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades, Paris, France
| | | | | | | | - Emilie Panafieu
- LEAT antenne Imagine- SFR Necker INSERM US 24, Paris, France
| | - Frank Blec
- LEAT antenne Imagine- SFR Necker INSERM US 24, Paris, France
| | - Mathieu Coureuil
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades, Paris, France
| | - Eric Frapy
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades, Paris, France
| | - Vassilis Tsatsaris
- Maternité Port-Royal, hôpital Cochin, GHU Centre Paris cité, AP-HP, Paris, France
- FHU PREMA, Maternité Port-Royal, Paris, France
| | - Stephane Bonacorsi
- IAME, UMR 1137, INSERM, Université Paris Cité, Paris, France
- Laboratoire de Microbiologie, Hôpital Robert Debré, AP-HP, Paris, France
| | - David Skurnik
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades, Paris, France.
- Department of Microbiology, Necker Hospital, University de Paris, Paris, France.
- FHU PREMA, Maternité Port-Royal, Paris, France.
| |
Collapse
|
24
|
Jennert F, Schaaf D, Nau R, Kohler TP, Hammerschmidt S, Häusler D, Valentin-Weigand P, Seele J. Hydrogen peroxide is responsible for the cytotoxic effects of Streptococcus pneumoniae on primary microglia in the absence of pneumolysin. J Innate Immun 2024; 16:000536514. [PMID: 38569474 PMCID: PMC11060703 DOI: 10.1159/000536514] [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: 03/03/2023] [Accepted: 01/25/2024] [Indexed: 04/05/2024] Open
Abstract
INTRODUCTION Streptococcus pneumoniae is the most common cause of bacterial meningitis and meningoencephalitis in humans. The bacterium produces numerous virulence determinants, among them hydrogen peroxide (H2O2) and pneumolysin (Ply), which contribute to bacterial cytotoxicity. Microglia, the resident phagocytes in the brain, are distinct from other macrophages, and we thus compared their susceptibility to pneumococcal toxicity and their ability to phagocytose pneumococci with those of bone marrow-derived macrophages (BMDM). METHODS Microglia and BMDM were co-incubated with S. pneumoniae D39 to analyze survival of phagocytes by fluorescence microscopy, bacterial growth by quantitative plating, and phagocytosis by an antibiotic protection assay. Ply was detected by hemolysis assay and Western blot analysis. RESULTS We found that microglia were killed during pneumococcal infection with a wild-type and an isogenic ply-deficient mutant, whereas viability of BMDM was not affected by pneumococci. Treatment with recombinant Ply showed a dose-dependent cytotoxic effect on microglia and BMDM. However, high concentrations of recombinant Ply were required and under the chosen experimental conditions, Ply was not detectable in the supernatant during infection of microglia. Inactivation of H2O2 by exogenously added catalase abolished its cytotoxic effect. Consequently, infection of microglia with pneumococci deficient for the pyruvate oxidase SpxB, primarily producing H2O2, resulted in reduced killing of microglia. CONCLUSION Taken together, in the absence of Ply, H2O2 caused cell death in primary phagocytes in concentrations produced by pneumococci.
Collapse
Affiliation(s)
- Franziska Jennert
- University of Veterinary Medicine Hannover, Institute for Microbiology, Center for Infection Medicine, Hannover, Germany
| | - Désirée Schaaf
- University of Veterinary Medicine Hannover, Institute for Microbiology, Center for Infection Medicine, Hannover, Germany
| | - Roland Nau
- University Medical Center Göttingen, Department of Neuropathology, Göttingen, Germany
- Evangelisches Krankenhaus Göttingen-Weende, Department of Geriatrics, Göttingen, Germany
| | - Thomas P. Kohler
- Greifswald University, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Greifswald, Germany
| | - Sven Hammerschmidt
- Greifswald University, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Greifswald, Germany
| | - Darius Häusler
- University Medical Center Göttingen, Department of Neuropathology, Göttingen, Germany
- Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany
| | - Peter Valentin-Weigand
- University of Veterinary Medicine Hannover, Institute for Microbiology, Center for Infection Medicine, Hannover, Germany
| | - Jana Seele
- University Medical Center Göttingen, Department of Neuropathology, Göttingen, Germany
- Evangelisches Krankenhaus Göttingen-Weende, Department of Geriatrics, Göttingen, Germany
| |
Collapse
|
25
|
Hasegawa H, Kiyofuji S, Umekawa M, Shinya Y, Okamoto K, Shono N, Kondo K, Shin M, Saito N. Profiles of central nervous system surgical site infections in endoscopic transnasal surgery exposing the intradural space. J Hosp Infect 2024; 146:166-173. [PMID: 37516279 DOI: 10.1016/j.jhin.2023.06.033] [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: 04/30/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/31/2023]
Abstract
OBJECTIVE Despite its efficacy and minimal invasiveness, the clean-contaminated nature of endoscopic transnasal surgery (ETS) may be susceptible to central nervous system surgical site infections (CNS-SSIs), especially when involving intradural exposure. However, the profiles of ETS-associated CNS-SSIs are not fully elucidated. METHODS The institutional ETS cases performed between May 2017 and March 2023 were retrospectively analysed. The incidences of CNS-SSIs were calculated, and their risk factors examined. RESULTS The incidence of CNS-SSIs was 2.3% (7/305) in the entire cohort and 5.0% (7/140) in ETSs with intradural exposure. All the CNS-SSIs were meningitis and developed following ETS with intradural exposure. The incidences were 0%, 5.6% and 5.8% in ETSs with Esposito grade 1, 2 and 3 intraoperative cerebrospinal fluid leakage, respectively. Among the pre- and intra-operative factors, body mass index (unit odds ratio (OR), 0.62; 95% confidence interval (CI), 0.44-0.89; P<0.01), serum albumin (unit OR, 0.03; 95% CI, 0.0007-0.92; P=0.02), and American Society of Anesthesiologists physical status score (unit OR, 20.7; 95% CI, 1.65-259; P<0.01) were significantly associated with CNS-SSIs. Moreover, postoperative cerebrospinal fluid leakage was also significantly associated with CNS-SSIs (OR, 18.4; 95% CI, 3.55-95.0; P<0.01). CONCLUSIONS The incidence of ETS-associated CNS-SSIs is acceptably low. Intradural exposure was a prerequisite for CNS-SSIs. Malnutrition and poor comorbidity status should be recognized as important risks for CNS-SSIs in ETS.
Collapse
Affiliation(s)
- H Hasegawa
- Department of Neurosurgery, University of Tokyo, Tokyo, Japan.
| | - S Kiyofuji
- Department of Neurosurgery, University of Tokyo, Tokyo, Japan
| | - M Umekawa
- Department of Neurosurgery, University of Tokyo, Tokyo, Japan
| | - Y Shinya
- Department of Neurosurgery, University of Tokyo, Tokyo, Japan
| | - K Okamoto
- Department of Infectious Diseases, University of Tokyo, Tokyo, Japan
| | - N Shono
- Department of Neurosurgery, University of Tokyo, Tokyo, Japan
| | - K Kondo
- Department of Otorhinolaryngology, University of Tokyo, Tokyo, Japan
| | - M Shin
- Department of Neurosurgery, Teikyo University, Tokyo, Japan
| | - N Saito
- Department of Neurosurgery, University of Tokyo, Tokyo, Japan
| |
Collapse
|
26
|
Gao S, Jin W, Quan Y, Li Y, Shen Y, Yuan S, Yi L, Wang Y, Wang Y. Bacterial capsules: Occurrence, mechanism, and function. NPJ Biofilms Microbiomes 2024; 10:21. [PMID: 38480745 PMCID: PMC10937973 DOI: 10.1038/s41522-024-00497-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
In environments characterized by extended multi-stress conditions, pathogens develop a variety of immune escape mechanisms to enhance their ability to infect the host. The capsules, polymers that bacteria secrete near their cell wall, participates in numerous bacterial life processes and plays a crucial role in resisting host immune attacks and adapting to their niche. Here, we discuss the relationship between capsules and bacterial virulence, summarizing the molecular mechanisms of capsular regulation and pathogenesis to provide new insights into the research on the pathogenesis of pathogenic bacteria.
Collapse
Affiliation(s)
- Shuji Gao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Wenjie Jin
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Yingying Quan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Yue Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Yamin Shen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Shuo Yuan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
| | - Li Yi
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China
- College of Life Science, Luoyang Normal University, Luoyang, 471934, China
| | - Yuxin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China.
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China.
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471000, China.
- Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, 471003, China.
| |
Collapse
|
27
|
Vollmuth N, Sin J, Kim BJ. Host-microbe interactions at the blood-brain barrier through the lens of induced pluripotent stem cell-derived brain-like endothelial cells. mBio 2024; 15:e0286223. [PMID: 38193670 PMCID: PMC10865987 DOI: 10.1128/mbio.02862-23] [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] [Indexed: 01/10/2024] Open
Abstract
Microbe-induced meningoencephalitis/meningitis is a life-threatening infection of the central nervous system (CNS) that occurs when pathogens are able to cross the blood-brain barrier (BBB) and gain access to the CNS. The BBB consists of highly specialized brain endothelial cells that exhibit specific properties to allow tight regulation of CNS homeostasis and prevent pathogen crossing. However, during meningoencephalitis/meningitis, the BBB fails to protect the CNS. Modeling the BBB remains a challenge due to the specialized characteristics of these cells. In this review, we cover the induced pluripotent stem cell-derived, brain-like endothelial cell model during host-pathogen interaction, highlighting the strengths and recent work on various pathogens known to interact with the BBB. As stem cell technologies are becoming more prominent, the stem cell-derived, brain-like endothelial cell model has been able to reveal new insights in vitro, which remain challenging with other in vitro cell-based models consisting of primary human brain endothelial cells and immortalized human brain endothelial cell lines.
Collapse
Affiliation(s)
- Nadine Vollmuth
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Jon Sin
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Brandon J. Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa, Alabama, USA
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, Alabama, USA
| |
Collapse
|
28
|
Sudo RYU, Câmara MCC, Kieling SV, Marques IR, Mesquita Y, Piepenbrink BE, Mari PC. Shorter versus longer duration of antibiotic treatment in children with bacterial meningitis: a systematic review and meta-analysis. Eur J Pediatr 2024; 183:61-71. [PMID: 37870611 DOI: 10.1007/s00431-023-05275-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 10/02/2023] [Accepted: 10/08/2023] [Indexed: 10/24/2023]
Abstract
The optimal duration of antibiotic treatment for the most common bacterial meningitis etiologies in the pediatric population, namely Streptococcus pneumoniae, Haemophilus influenzae, and Neisseria meningitidis, is not well-established in the literature. Therefore, we aimed to perform an updated meta-analysis comparing shorter versus longer antibiotic treatment in children with meningitis. PubMed, EMBASE, and Cochrane databases were searched for randomized controlled trials (RCTs) that compared shorter (up to 7 days) versus longer (10 days or double the days of the equivalent short course) duration of antibiotic treatment in children with meningitis and reported the outcomes of treatment failure, death, neurologic sequelae, non-neurologic complications, hearing impairment, nosocomial infection, and relapse. Heterogeneity was examined with I2 statistics. RevMan 5.4.1 was used for statistical analysis and RoB-2 (Cochrane) for risk of bias assessment. Of 684 search results, 6 RCTs were included, with a cohort of 1333 children ages 3 weeks to 15.5 years, of whom 49.51% underwent a short antibiotic course. All RCTs included monotherapy with ceftriaxone, except one, which added vancomycin as well. No differences were found comparing the short and long duration of therapy concerning treatment failure, relapse, mortality, and neurologic complications at discharge and at follow-up. Conclusion: Because no statistically significant differences were found between groups for the analyzed outcomes, the results of this meta-analysis support shorter therapy. However, generalizing these results to complicated meningitis and infections caused by other pathogens should be made with caution. (PROSPERO identifier: CRD42022369843). What is Known: • Current recommendations on the duration of antibiotic therapy for bacterial meningitis are mostly based on clinical practice. • Defining an optimal duration of antibiotic therapy is essential for antimicrobial stewardship achievement, improving patient outcomes, and minimizing adverse effects. What is New: • There are no differences between shorter versus longer antibiotic treatment duration in regard to treatment failure, relapse, mortality, neurologic complications, and hearing impairment at discharge and at follow-up.
Collapse
Affiliation(s)
- Renan Yuji Ura Sudo
- Division of Medicine, Federal University of Grande Dourados, Dourados, MS, Brazil.
| | | | | | - Isabela Reis Marques
- Division of Medicine, Universitat Internacional de Catalunya, Barcelona, CAT, Spain
| | - Yasmin Mesquita
- Division of Medicine, Federal University of Rio de Janeiro, Macaé, RJ, Brazil
| | - Blake Earl Piepenbrink
- Division of Internal Medicine, Primary Care, University of Connecticut, Farmington, CT, USA
| | - Paula Chaves Mari
- Pediatric Division, Cleveland Clinic Children's Hospital, Cleveland, OH, USA
| |
Collapse
|
29
|
Generoso JS, Faller CJ, Collodel A, Catalão CHR, Dominguini D, Petronilho F, Barichello T, Giridharan VV. NLRP3 Activation Contributes to Memory Impairment in an Experimental Model of Pneumococcal Meningitis. Mol Neurobiol 2024; 61:239-251. [PMID: 37603152 PMCID: PMC11409915 DOI: 10.1007/s12035-023-03549-8] [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: 05/08/2023] [Accepted: 07/25/2023] [Indexed: 08/22/2023]
Abstract
Bacterial meningitis is considered a life-threatening condition with high mortality rates. In response to the infection, signaling cascades, producing pro-inflammatory mediators trigger an exacerbated host immune response. Another inflammatory pathway occurs through the activation of inflammasomes. Studies highlight the role of the NLR family pyrin domain containing 3 (NLRP3) in central nervous system disorders commonly involved in neuroinflammation. We aimed to investigate the role of NLRP3 and its inhibitor MCC950 on neurochemical, immunological, and behavioral parameters in the early and late stages of experimental pneumococcal meningitis. For this, adult male Wistar rats received an intracisternal injection of Streptococcus pneumoniae or artificial cerebrospinal fluid as a placebo. The animals were divided into control/saline, control/MCC950, meningitis/saline, and meningitis/MCC950. Immediately after the meningitis induction, the animals received 140 ng/kg MCC950 via intracisternal injection. For the acute protocol, 24 h after induction, brain structures were collected to evaluate cytokines, NLRP3, and microglia. In the long-term group, the animals were submitted to open field and recognition of new objects tests at ten days after the meningitis induction. After the behavioral tests, the same markers were evaluated. The animals in the meningitis group at 24 h showed increased levels of cytokines, NLRP3, and IBA-1 expression, and the use of the MCC950 significantly reduced those levels. Although free from infection, ten days after meningitis induction, the animals in the meningitis group had elevated cytokine levels and demonstrated behavioral deficits; however, the single dose of NLRP3 inhibitor rescued the behavior deficits and decreased the brain inflammatory profile.
Collapse
Affiliation(s)
- Jaqueline S Generoso
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| | - Cristiano Julio Faller
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Allan Collodel
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Carlos Henrique Rocha Catalão
- Faillace Department of Psychiatry and Behavioral Sciences, Translational Psychiatry Program, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Diogo Dominguini
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Tatiana Barichello
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Faillace Department of Psychiatry and Behavioral Sciences, Translational Psychiatry Program, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Vijayasree V Giridharan
- Faillace Department of Psychiatry and Behavioral Sciences, Translational Psychiatry Program, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
| |
Collapse
|
30
|
Alanazi F, Raghunandanan S, Priya R, Yang XF. The Rrp2-RpoN-RpoS pathway plays an important role in the blood-brain barrier transmigration of the Lyme disease pathogen. Infect Immun 2023; 91:e0022723. [PMID: 37874144 PMCID: PMC10652863 DOI: 10.1128/iai.00227-23] [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: 06/13/2023] [Accepted: 09/14/2023] [Indexed: 10/25/2023] Open
Abstract
Lyme disease, caused by Borrelia (or Borreliella) burgdorferi, is a complex multisystemic disorder that includes Lyme neuroborreliosis resulting from the invasion of both the central and peripheral nervous systems. However, factors that enable the pathogen to cross the blood-brain barrier (BBB) and invade the central nervous system (CNS) are still not well understood. The objective of this study was to identify the B. burgdorferi factors required for BBB transmigration. We utilized a transwell BBB model based on human brain-microvascular endothelial cells and focused on investigating the Rrp2-RpoN-RpoS pathway, a central regulatory pathway that is essential for mammalian infection by B. burgdorferi. Our results demonstrated that the Rrp2-RpoN-RpoS pathway is crucial for BBB transmigration. Furthermore, we identified OspC, a major surface lipoprotein controlled by the Rrp2-RpoN-RpoS pathway, as a significant contributor to BBB transmigration. Constitutive production of OspC in a mutant defective in the Rrp2-RpoN-RpoS pathway did not rescue the impairment in BBB transmigration, indicating that this pathway controls additional factors for this process. Two other major surface lipoproteins controlled by this pathway, DbpA/B and BBK32, appeared to be dispensable for BBB transmigration. In addition, both the surface lipoprotein OspA and the Rrp1 pathway, which are required B. burgdorferi colonization in the tick vector, were found not required for BBB transmigration. Collectively, our findings using in vitro transwell assays uncover another potential role of the Rrp2-RpoN-RpoS pathway in BBB transmigration of B. burgdorferi and invasion into the CNS.
Collapse
Affiliation(s)
- Fuad Alanazi
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Sajith Raghunandanan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Raj Priya
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - X. Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
31
|
Fohmann I, Weinmann A, Schumacher F, Peters S, Prell A, Weigel C, Spiegel S, Kleuser B, Schubert-Unkmeir A. Sphingosine kinase 1/S1P receptor signaling axis is essential for cellular uptake of Neisseria meningitidis in brain endothelial cells. PLoS Pathog 2023; 19:e1011842. [PMID: 38033162 PMCID: PMC10715668 DOI: 10.1371/journal.ppat.1011842] [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: 07/29/2023] [Revised: 12/12/2023] [Accepted: 11/18/2023] [Indexed: 12/02/2023] Open
Abstract
Invasion of brain endothelial cells (BECs) is central to the pathogenicity of Neisseria meningitidis infection. Here, we established a key role for the bioactive sphingolipid sphingosine-1-phosphate (S1P) and S1P receptor (S1PR) 2 in the uptake process. Quantitative sphingolipidome analyses of BECs infected with N. meningitidis revealed elevated S1P levels, which could be attributed to enhanced expression of the enzyme sphingosine kinase 1 and its activity. Increased activity was dependent on the interaction of meningococcal type IV pilus with the endothelial receptor CD147. Concurrently, infection led to increased expression of the S1PR2. Blocking S1PR2 signaling impaired epidermal growth factor receptor (EGFR) phosphorylation, which has been shown to be involved in cytoskeletal remodeling and bacterial endocytosis. Strikingly, targeting S1PR1 or S1PR3 also interfered with bacterial uptake. Collectively, our data support a critical role of the SphK/S1P/S1PR axis in the invasion of N. meningitidis into BECs, defining a potential target for adjuvant therapy.
Collapse
Affiliation(s)
- Ingo Fohmann
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Alina Weinmann
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Fabian Schumacher
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Simon Peters
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Agata Prell
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Cynthia Weigel
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States of America
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, United States of America
| | - Burkhard Kleuser
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany
| | | |
Collapse
|
32
|
Kalchev Y, Argirova P, Boev I, Yaneva A, Vatev N, Stoycheva M, Murdjeva M. Cytokine profile in patients with acute bacterial meningitis. Cytokine 2023; 170:156315. [PMID: 37544134 DOI: 10.1016/j.cyto.2023.156315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Bacterial meningitis is a life-threatening disease with high mortality and common long-term sequelae. The inflammatory response in the subarachnoid space, modulated by different cytokines, plays a major role in the pathogenesis of acute central nervous system infections. We aimed to examine correlations of interleukin (IL)-6, IL-8, IL-10, IL-12(p40), and tumor necrosis factor (TNF)-α levels with disease severity, complications, and outcome in patients with acute bacterial meningitis. METHODS The study involved 30 patients with bacterial meningitis/meningoencephalitis admitted to the University Hospital St. George, Plovdiv over a period of 4 years. Patients were selected based on clinical presentation and laboratory abnormalities, consistent with a neuroinfection. Enzyme-linked immunosorbent assay was used to measure the studied cytokines in both cerebrospinal fluid (CSF) and serum in parallel. For microbiological diagnosis multiplex, polymerase chain reaction, and CSF culture were used. RESULTS In patients with acute bacterial meningitis CSF levels of IL-6, IL-8, IL-10, and TNF-α are significantly increased than in serum. CSF TNF-α, CSF IL-8, and CSF IL-10 had a moderate negative correlation to CSF glucose. It was found that serum IL-8 is significantly elevated in patients who experienced neurological complications, have severe clinical course, and in deceased patients. CSF IL-10 is increased only in patients with severe acute bacterial meningitis. CONCLUSION Among patients with acute bacterial meningitis serum IL-8 could delineate these with increased risk of neurological complications, severe clinical course, and fatal outcome. Serum IL-8 and CSF IL-10 could be used as indicators of disease severity.
Collapse
Affiliation(s)
- Y Kalchev
- Department of Medical Microbiology and Immunology "Prof. Dr. Elissay Yanev", Faculty of Pharmacy, Medical University - Plovdiv, Bulgaria; Laboratory of Microbiology, University Hospital St. George, Plovdiv, Bulgaria; Research Institute at Medical University - Plovdiv, Bulgaria.
| | - P Argirova
- Department of Infectious Diseases and Parasitology, Faculty of Medicine, Medical University - Plovdiv, Bulgaria
| | - I Boev
- Department of Infectious Diseases and Parasitology, Faculty of Medicine, Medical University - Plovdiv, Bulgaria
| | - A Yaneva
- Department of Medical Informatics, Biostatistics, and eLearning, Faculty of Public Health, Medical University - Plovdiv, Bulgaria
| | - N Vatev
- Department of Epidemiology and Disaster Medicine, Faculty of Public Health, Medical University - Plovdiv, Bulgaria
| | - M Stoycheva
- Department of Infectious Diseases and Parasitology, Faculty of Medicine, Medical University - Plovdiv, Bulgaria; Clinic of Infectious Diseases, University Hospital St. George, Plovdiv, Bulgaria
| | - M Murdjeva
- Department of Medical Microbiology and Immunology "Prof. Dr. Elissay Yanev", Faculty of Pharmacy, Medical University - Plovdiv, Bulgaria; Laboratory of Microbiology, University Hospital St. George, Plovdiv, Bulgaria; Research Institute at Medical University - Plovdiv, Bulgaria
| |
Collapse
|
33
|
Cutright AJ, Al Mohanna T, Matthews EL, Aulds JM, Thornton JA, Stokes SL, Emerson JP. Calorimetric analysis of AdcR and its interactions with zinc(II) and DNA. J Inorg Biochem 2023; 247:112305. [PMID: 37441924 DOI: 10.1016/j.jinorgbio.2023.112305] [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: 04/18/2023] [Revised: 06/09/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023]
Abstract
Zinc(II) ions play critical roles in all known life as structurally important stabilizing ions in proteins, catalytically active metals in enzymes, and signaling agents impacting physiological changes. To maintain homeostasis, the intracellular concentration of zinc(II) is strictly controlled by a family of metal-regulatory proteins in both prokaryotic and eukaryotic organisms. In S. pneumoniae, there are two proteins that share responsibility for Zn2+ homeostasis, one of them is the Adhesin Competence Repressor (AdcR) and it binds to a specific double-stranded DNA binding domain (dsDNA). AdcR has been structurally characterized containing two zinc(II) metal centers per monomeric unit. Here we report data collected from differential scanning calorimetry (DSC) experiments aimed to measure the structural stability of AdcR, the fully complimented Zn2AdcR complex, and the protein/DNA complex Zn2AdcR/dsDNA. Thermograms collected from DSC experiments yielded endothermic unfolding events for AdcR, Zn2AdcR, and Zn2AdcR/dsDNA complex at 55.6, 70.2, and 56.6 °C, respectively. A non-two state unfolding model best fits the data, giving ΔH terms associated with these thermal unfolding events of 5.1, 7.1, and 4.9 kcal/mol. These data allow for the development of a thermodynamic cycle connecting both zinc(II) and DNA binding to AdcR. Furthermore, pairing this newly reported data with known association constants for zinc(II) and DNA binding allowed for the generation of thermodynamic profiles for both zinc(II) binding to AdcR and Zn2AdcR binding to DNA, which show both are decisively entropy-driven processes.
Collapse
Affiliation(s)
- Alexander J Cutright
- Department of Chemistry, Mississippi State University, Mississippi State, MS 39762, United States
| | - Thualfeqar Al Mohanna
- Department of Chemistry, Mississippi State University, Mississippi State, MS 39762, United States
| | - Erin L Matthews
- Department of Chemistry, Mississippi State University, Mississippi State, MS 39762, United States
| | - James M Aulds
- Department of Chemistry, Mississippi State University, Mississippi State, MS 39762, United States
| | - Justin A Thornton
- Department of Biological Sciences, Mississippi State University, Mississippi State, MS 39762, United States
| | - Sean L Stokes
- Department of Chemistry, Mississippi State University, Mississippi State, MS 39762, United States
| | - Joseph P Emerson
- Department of Chemistry, Mississippi State University, Mississippi State, MS 39762, United States.
| |
Collapse
|
34
|
Cheng Z, Zheng Y, Yang W, Sun H, Zhou F, Huang C, Zhang S, Song Y, Liang Q, Yang N, Li M, Liu B, Feng L, Wang L. Pathogenic bacteria exploit transferrin receptor transcytosis to penetrate the blood-brain barrier. Proc Natl Acad Sci U S A 2023; 120:e2307899120. [PMID: 37733740 PMCID: PMC10523449 DOI: 10.1073/pnas.2307899120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/23/2023] [Indexed: 09/23/2023] Open
Abstract
The human blood-brain barrier (BBB) comprises a single layer of brain microvascular endothelial cells (HBMECs) protecting the brain from bloodborne pathogens. Meningitis is among the most serious diseases, but the mechanisms by which major meningitis-causing bacterial pathogens cross the BBB to reach the brain remain poorly understood. We found that Streptococcus pneumoniae, group B Streptococcus, and neonatal meningitis Escherichia coli commonly exploit a unique vesicle fusion mechanism to hitchhike on transferrin receptor (TfR) transcytosis to cross the BBB and illustrated the details of this process in human BBB model in vitro and mouse model. Toll-like receptor signals emanating from bacteria-containing vesicles (BCVs) trigger K33-linked polyubiquitination at Lys168 and Lys181 of the innate immune regulator TRAF3 and then activate the formation of a protein complex containing the guanine nucleotide exchange factor RCC2, the small GTPase RalA and exocyst subcomplex I (SC I) on BCVs. The distinct function of SEC6 in SC I, interacting directly with RalA on BCVs and the SNARE protein SNAP23 on TfR vesicles, tethers these two vesicles and initiates the fusion. Our results reveal that innate immunity triggers a unique modification of TRAF3 and the formation of the HBMEC-specific protein complex on BCVs to authenticate the precise recognition and selection of TfR vesicles to fuse with and facilitate bacterial penetration of the BBB.
Collapse
Affiliation(s)
- Zhihui Cheng
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin300071, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin300071, China
| | - Yangyang Zheng
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin300071, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin300457, China
| | - Wen Yang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin300071, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin300457, China
| | - Hongmin Sun
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin300071, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin300457, China
| | - Fangyu Zhou
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin300071, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin300457, China
| | - Chuangjie Huang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin300071, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin300457, China
| | - Shuwen Zhang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin300071, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin300457, China
| | - Yingying Song
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin300071, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin300457, China
| | - Qi’an Liang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin300071, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin300071, China
| | - Nan Yang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin300071, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin300071, China
| | - Meifang Li
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin300071, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin300071, China
| | - Bin Liu
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin300071, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin300457, China
| | - Lu Feng
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin300071, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin300457, China
| | - Lei Wang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin300071, China
- TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin300457, China
| |
Collapse
|
35
|
Ayoub MA. Hijacking of GPCRs and RTKs by pathogens. Cell Signal 2023:110802. [PMID: 37437829 DOI: 10.1016/j.cellsig.2023.110802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/29/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
Pathogens exploit multiple cellular and molecular pathways in the host organisms for their entry, survival and dissemination. The cell surface receptors such as G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs) constitute the targets of many pathogens. This is due to the ubiquitous expression of these two receptor families in the organism and their pivotal role in various cellular and physiological processes. At the molecular level, receptor hijacking implies either direct or indirect interactions between pathogens' effectors or toxins with GPCRs and RTKs at the cell surface thereby interfering with their activation and their downstream signaling pathways inside the host cells. As a result, the pathogens manipulate and redirect GPCR/RTK-mediated signaling pathways and different aspects of cell function for their benefit. The review presents a compilation of the major examples of pathogen infections where GPCRs and RTKs and their related intracellular signaling pathways are targeted. This provides a molecular basis for pathogens hijacking cell signaling and their virulence. Our understanding of such complex host-pathogen interactions at the molecular level will open new opportunities to develop new prophylactic and therapeutic approaches against infections. In this context, the pharmacological targeting of GPCRs and RTKs may be a promising approach.
Collapse
Affiliation(s)
- Mohammed Akli Ayoub
- Biology Department, College of Arts and Sciences, Khalifa University, PO Box 127788, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
36
|
Antoine C, Laforêt F, Goya-Jorge E, Gonza I, Lebrun S, Douny C, Duprez JN, Fall A, Taminiau B, Scippo ML, Daube G, Thiry D, Delcenserie V. Phage Targeting Neonatal Meningitis E. coli K1 In Vitro in the Intestinal Microbiota of Pregnant Donors and Impact on Bacterial Populations. Int J Mol Sci 2023; 24:10580. [PMID: 37445758 DOI: 10.3390/ijms241310580] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Escherichia coli K1 is a leading cause of neonatal meningitis. The asymptomatic carriage of these strains in the maternal intestinal microbiota constitutes a risk of vertical transmission to the infant at birth. The aim of this work was to evaluate the efficacy of phage therapy against E. coli K1 in an intestinal environment and its impact on the intestinal microbiota. For this purpose, three independent experiments were conducted on the SHIME® system, the first one with only the phage vB_EcoP_K1_ULINTec4, the second experiment with only E. coli K1 and the last experiment with both E. coli K1 and the phage. Microbiota monitoring was performed using metagenetics, qPCR, SCFA analysis and the induction of AhR. The results showed that phage vB_EcoP_K1_ULINTec4, inoculated alone, was progressively cleared by the system and replicates in the presence of its host. E. coli K1 persisted in the microbiota but decreased in the presence of the phage. The impact on the microbiota was revealed to be donor dependent, and the bacterial populations were not dramatically affected by vB_K1_ULINTec4, either alone or with its host. In conclusion, these experiments showed that the phage was able to infect the E. coli K1 in the system but did not completely eliminate the bacterial load.
Collapse
Affiliation(s)
- Céline Antoine
- Laboratory of Veterinary Bacteriology, Department of Infectious and Parasitic Diseases, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
- Laboratory of Food Quality Management, Food Science Department, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Fanny Laforêt
- Laboratory of Veterinary Bacteriology, Department of Infectious and Parasitic Diseases, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
- Laboratory of Food Quality Management, Food Science Department, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Elizabeth Goya-Jorge
- Laboratory of Food Quality Management, Food Science Department, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Irma Gonza
- Laboratory of Food Quality Management, Food Science Department, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Sarah Lebrun
- Laboratory of Food Quality Management, Food Science Department, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Caroline Douny
- Laboratory of Food Analysis, Department of Food Sciences, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Jean-Noël Duprez
- Laboratory of Veterinary Bacteriology, Department of Infectious and Parasitic Diseases, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Abdoulaye Fall
- FoodChain ID Genomics, En Hayeneux 62, 4040 Herstal, Belgium
| | - Bernard Taminiau
- Laboratory of Microbiology, Department of Food Sciences, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Marie-Louise Scippo
- Laboratory of Food Analysis, Department of Food Sciences, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Georges Daube
- Laboratory of Microbiology, Department of Food Sciences, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Damien Thiry
- Laboratory of Veterinary Bacteriology, Department of Infectious and Parasitic Diseases, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Véronique Delcenserie
- Laboratory of Food Quality Management, Food Science Department, FARAH and Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| |
Collapse
|
37
|
Qu X, Dou B, Yang R, Tan C, Chen H, Wang X. C-X-C Motif Chemokine 3 Promotes the Inflammatory Response of Microglia after Escherichia coli-Induced Meningitis. Int J Mol Sci 2023; 24:10432. [PMID: 37445610 DOI: 10.3390/ijms241310432] [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: 05/27/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Meningitis is a major clinical manifestation of Escherichia coli (E. coli) infection characterized by inflammation of the meninges and subarachnoid space. Many chemokines are secreted during meningitic E. coli infection, of which C-X-C motif chemokine 3 (CXCL3) is the most highly expressed. However, it is unclear how CXCL3 plays a role in meningitic E. coli infection. Therefore, this study used in vitro and in vivo assays to clarify these contributions and to identify novel therapeutic targets for central nervous system inflammation. We found a significantly upregulated expression of CXCL3 in human brain microvascular endothelial cells and U251 cells after meningitic E. coli infection, and the CXCL3 receptor, C-X-C motif chemokine receptor 2 (CXCR2), was expressed in microglia. Furthermore, CXCL3 induced M1 microglia by selectively activating mitogen-activated protein kinases signaling and significantly upregulating tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, nitric oxide synthase 2 (NOS2), and cluster of differentiation 86 (CD86) expression levels, promoting an inflammatory response. Our findings clarify the role of CXCL3 in meningitic E. coli-induced neuroinflammation and demonstrate that CXCL3 may be a potential therapeutic target for future investigation and prevention of E. coli-induced neuroinflammation.
Collapse
Affiliation(s)
- Xinyi Qu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan 430070, China
| | - Beibei Dou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan 430070, China
| | - Ruicheng Yang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan 430070, China
| | - Chen Tan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| | - Xiangru Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan 430070, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan 430070, China
| |
Collapse
|
38
|
Guo H, Jin W, Liu K, Liu S, Mao S, Zhou Z, Xie L, Wang G, Chen Y, Liang Y. Oral GSH Exerts a Therapeutic Effect on Experimental Salmonella Meningitis by Protecting BBB Integrity and Inhibiting Salmonella-induced Apoptosis. J Neuroimmune Pharmacol 2023; 18:112-126. [PMID: 36418663 DOI: 10.1007/s11481-022-10055-6] [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: 05/18/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022]
Abstract
Bacterial meningitis (BM) is the main cause of the central nervous system (CNS) infection and continues to be an important cause of mortality and morbidity. Glutathione (GSH), an endogenous tripeptide antioxidant, has been proved to exert crucial role in reducing superoxide radicals, hydroxyl radicals and peroxynitrites. The purpose of this study is to expand the application scope of GSH via exploring its therapeutic effect on BM caused by Salmonella typhimurium SL1344 and then provide a novel approach for the treatment of BM. The results suggested that intragastric administration of GSH could significantly increase median survival and improve experimental autoimmune encephalomyelitis score of BM model mice. However, exogenous GSH did not affect the adhesion, invasion and cytotoxicity of SL1344 to C6, BV2 and primary microglia. Due to the contradiction between the therapeutic and bactericidal effects of GSH, the effect of GSH on blood-brain barrier (BBB) was investigated to explore its action target for the treatment of meningitis. GSH was found to repair the damage of BBB and then prevent the leakage of SL1344 from the brain to the blood circulation. The repaired BBB could also effectively reduce the entry of macrophages and neutrophils into the brain, and significantly reverse the microglia activation induced by SL1344. More importantly, exogenous GSH was proved to reduce mouse brain cell apoptosis by inhibiting the activation of caspase-8 followed by caspase-3, and reversing the up-regulation of ICAD and PARP-1 caused by SL1344.
Collapse
Affiliation(s)
- Huimin Guo
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, 210009, Nanjing, P.R. China
| | - Wei Jin
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, 210009, Nanjing, P.R. China
| | - Keanqi Liu
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, 210009, Nanjing, P.R. China
| | - Shijia Liu
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, 210009, Nanjing, P.R. China
| | - Shuying Mao
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, 210009, Nanjing, P.R. China
| | - Zhihao Zhou
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, 210009, Nanjing, P.R. China
| | - Lin Xie
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, 210009, Nanjing, P.R. China
| | - Guangji Wang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, 210009, Nanjing, P.R. China.
| | - Yugen Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Road, Qinhuai District, 210000, Nanjing, P.R. China.
| | - Yan Liang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang 24, 210009, Nanjing, P.R. China.
| |
Collapse
|
39
|
Caragheorgheopol R, Țucureanu C, Lazăr V, Florescu SA, Lazăr DS, Caraş I. Cerebrospinal fluid cytokines and chemokines exhibit distinct profiles in bacterial meningitis and viral meningitis. Exp Ther Med 2023; 25:204. [PMID: 37090083 PMCID: PMC10119981 DOI: 10.3892/etm.2023.11903] [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: 09/20/2022] [Accepted: 02/24/2023] [Indexed: 04/25/2023] Open
Abstract
Differential diagnosis of bacterial meningitis (BM) and viral meningitis (VM) is a critical clinical challenge, as the early and accurate identification of the causative agent determines the appropriate treatment regimen and markedly improves patient outcomes. Clinical and experimental studies have demonstrated that the pathogen and the host immune response contribute to mortality and neurological sequelae. As BM is associated with the activation of an inflammatory cascade, the patterns of pro- and anti-inflammatory cytokines/chemokines (CTs/CKs) present in the cerebrospinal fluid (CSF) in response to the immune assault may be useful as sensitive markers for differentiating BM from VM. In the present study, the ability of CTs/CKs in the CSF to differentiate between BM and VM was investigated. For this, biochemical markers and CT/CK profiles were analysed in 145 CSF samples, divided into three groups: BM (n=61), VM (n=58) and the control group (C; n=26) comprising patients with meningism. The CSF concentrations of monocyte chemoattractant protein-1, interleukin (IL)-8, IL-1β, IL-6, macrophage inflammatory protein-1α (MIP-1α), epithelial-neutrophil activating peptide, IL-10, tumour necrosis factor-α (TNF-α), proteins and white blood cells were significantly higher and the CSF glucose level was significantly lower in the BM group compared with the VM and C groups (P<0.01). Correlation analysis identified 28 significant correlations between various CTs/CKs in the BM group (P<0.01), with the strongest positive correlations being for TNF-α/IL-6 (r=0.75), TNF-α/MIP-1α (r=0.69), TNF-α/IL-1β (r=0.64) and IL-1β/MIP-1α (r=0.64). To identify the optimum CT/CK patterns for predicting and classifying BM and VM, a dataset of 119 BM and VM samples was divided into training (n=90) and testing (n=29) subsets for use as input for a Random Forest (RF) machine learning algorithm. For the 29 test samples (15 BM and 14 VM), the RF algorithm correctly classified 28 samples, with 92% sensitivity and 93% specificity. The results show that the patterns of CT/CK levels in the CSF can be used to aid discrimination of BM and VM.
Collapse
Affiliation(s)
- Ramona Caragheorgheopol
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest 77206, Romania
- Immunology Laboratory, ‘Cantacuzino’ National Institute for Medico-Military Research and Development, Bucharest 050096, Romania
- Correspondence to: Mrs. Ramona Caragheorgheopol, Immunology Laboratory, ‘Cantacuzino’ National Institute for Medico-Military Research and Development, 103 Splaiul Independentei, Bucharest 050096, Romania
| | - Cătălin Țucureanu
- Immunology Laboratory, ‘Cantacuzino’ National Institute for Medico-Military Research and Development, Bucharest 050096, Romania
| | - Veronica Lazăr
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest 77206, Romania
| | - Simin Aysel Florescu
- Infectious Diseases Department II, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest 050474, Romania
- Clinical Department A5 for Infectious and Tropical Diseases, ‘Dr Victor Babes’ Clinical Hospital for Infectious and Tropical Diseases, Bucharest 030303, Romania
| | - Dragoş Stefan Lazăr
- Infectious Diseases Department II, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest 050474, Romania
- Adults Department B2, ‘Dr Victor Babes’ Clinical Hospital for Infectious and Tropical Diseases, Bucharest 030303, Romania
| | - Iuliana Caraş
- Immunology Laboratory, ‘Cantacuzino’ National Institute for Medico-Military Research and Development, Bucharest 050096, Romania
| |
Collapse
|
40
|
Herold R, Denzer L, Muranyi W, Stump-Guthier C, Ishikawa H, Schroten H, Schwerk C. The phosphoproteome of choroid plexus epithelial cells following infection with Neisseria meningitidis. Front Cell Infect Microbiol 2023; 13:1113528. [PMID: 37065199 PMCID: PMC10102474 DOI: 10.3389/fcimb.2023.1113528] [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: 12/01/2022] [Accepted: 03/03/2023] [Indexed: 04/03/2023] Open
Abstract
The Gram-negative bacterium Neisseria meningitidis, which causes meningitis in humans, has been demonstrated to manipulate or alter host signalling pathways during infection of the central nervous system (CNS). However, these complex signalling networks are not completely understood. We investigate the phosphoproteome of an in vitro model of the blood-cerebrospinal fluid barrier (BCSFB) based on human epithelial choroid plexus (CP) papilloma (HIBCPP) cells during infection with the N. meningitidis serogroup B strain MC58 in presence and absence of the bacterial capsule. Interestingly, our data demonstrates a stronger impact on the phosphoproteome of the cells by the capsule-deficient mutant of MC58. Using enrichment analyses, potential pathways, molecular processes, biological processes, cellular components and kinases were determined to be regulated as a consequence of N. meningitidis infection of the BCSFB. Our data highlight a variety of protein regulations that are altered during infection of CP epithelial cells with N. meningitidis, with the regulation of several pathways and molecular events only being detected after infection with the capsule-deficient mutant. Mass spectrometry proteomics data are available via ProteomeXchange with identifier PXD038560.
Collapse
Affiliation(s)
- Rosanna Herold
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lea Denzer
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Walter Muranyi
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carolin Stump-Guthier
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hiroshi Ishikawa
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Horst Schroten
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christian Schwerk
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
41
|
OmpA is involved in the invasion of duck brain microvascular endothelial cells by Riemerella anatipestifer. Vet Microbiol 2023; 280:109692. [PMID: 36863175 DOI: 10.1016/j.vetmic.2023.109692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 11/25/2022] [Accepted: 02/15/2023] [Indexed: 02/18/2023]
Abstract
Bacterial meningitis is a major cause of morbidity and mortality. Despite advances in antimicrobial chemotherapy, the disease remains detrimental to humans, livestock, and poultry. Riemerella anatipestifer is a gram-negative bacterium causing duckling serositis and meningitis. However, the virulence factors contributing to its binding and invasion of duck brain microvascular endothelial cells (DBMECs) and penetration of the blood-brain barrier (BBB) have never been reported. In this study, immortalized DBMECs were successfully generated and used as an in vitro-model of duck BBB. Furthermore, ompA gene deletion mutant of the pathogen and multiple complemented strains carrying the complete ompA gene and its truncated forms were constructed. Bacterial growth, invasion, and adhesion assays and animal experiments were performed. The results show that the OmpA protein of R. anatipestifer had no effect on bacterial growth and adhesion ability to DBMECs. The role of OmpA in the invasion of R. anatipestifer into DBMECs and duckling BBB was confirmed. The amino acids 230-242 of OmpA represents a key domain involved in R. anatipestifer invasion. In addition, another OmpA1164 protein constituted by the amino acids 102-488 within OmpA could function as a complete OmpA. The signal peptide sequence from amino acids 1-21 had no significant effect on OmpA functions. In conclusion, this study illustrated that OmpA is an important virulence factor mediating R. anatipestifer invasion of DBMECs and penetration of the duckling BBB.
Collapse
|
42
|
Younger DS. Pediatric neuropsychiatric disorders with motor and nonmotor phenomena. HANDBOOK OF CLINICAL NEUROLOGY 2023; 196:367-387. [PMID: 37620079 DOI: 10.1016/b978-0-323-98817-9.00028-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
The concept of pediatric autoimmune neuropsychiatric disorders associated with group A beta-hemolytic streptococcus (PANDAS) has become seminal since first introduced more than two decades ago. At the time of this writing, most neurologists, pediatricians, psychiatrists, and general pediatricians will probably have heard of this association or treated an affected child with PANDAS. The concept of an acute-onset, and typically self-limited, postinfectious autoimmune neuropsychiatric disorder resembling PANDAS manifesting vocal and motor tics and obsessive-compulsive disorder has broadened to other putative microbes and related endogenous and exogenous disease triggers. These disorders with common features of hypometabolism in the medial temporal lobe and hippocampus in brain 18fluorodeoxyglucose positron emission tomography fused to magnetic resonance imaging (FDG PET-MRI), form a spectrum: with the neuropsychiatric disorder Tourette syndrome and PANDAS with its well-defined etiopathogenesis at one end, and pediatric abrupt-onset neuropsychiatric syndrome (PANS), alone or associated with specific bacterial and viral pathogens, at the other end. The designation of PANS in the absence of a specific trigger, as an exclusionary diagnosis, reflects the current problem in nosology.
Collapse
Affiliation(s)
- David S Younger
- Department of Clinical Medicine and Neuroscience, CUNY School of Medicine, New York, NY, United States; Department of Medicine, Section of Internal Medicine and Neurology, White Plains Hospital, White Plains, NY, United States.
| |
Collapse
|
43
|
Zhang XW, An MX, Huang ZK, Ma L, Zhao D, Yang Z, Shi JX, Liu DX, Li Q, Wu AH, Chen YH, Zhao WD. Lpp of Escherichia coli K1 inhibits host ROS production to counteract neutrophil-mediated elimination. Redox Biol 2022; 59:102588. [PMID: 36592568 PMCID: PMC9823224 DOI: 10.1016/j.redox.2022.102588] [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: 11/20/2022] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 12/27/2022] Open
Abstract
Escherichia coli (E. coli) is the most common Gram-negative bacterial organism causing neonatal meningitis. The pathogenesis of E. coli meningitis, especially how E. coli escape the host immune defenses, remains to be clarified. Here we show that deletion of bacterial Lpp encoding lipoprotein significantly reduces the pathogenicity of E. coli K1 to induce high-degree of bacteremia necessary for meningitis. The Lpp-deleted E. coli K1 is found to be susceptible to the intracellular bactericidal activity of neutrophils, without affecting the release of neutrophil extracellular traps. The production of reactive oxygen species (ROS), representing the primary antimicrobial mechanism in neutrophils, is significantly increased in response to Lpp-deleted E. coli. We find this enhanced ROS response is associated with the membrane translocation of NADPH oxidase p47phox and p67phox in neutrophils. Then we constructed p47phox knockout mice and we found the incidence of bacteremia and meningitis in neonatal mice induced by Lpp-deleted E. coli is significantly recovered by p47phox knockout. Proteomic profile analysis show that Lpp deficiency induces upregulation of flagellar protein FliC in E. coli. We further demonstrate that FliC is required for the ROS induction in neutrophils by Lpp-deleted E. coli. Taken together, these data uncover the novel role of Lpp in facilitating intracellular survival of E. coli K1 within neutrophils. It can be inferred that Lpp of E. coli K1 is able to suppress FliC expression to restrain the activation of NADPH oxidase in neutrophils resulting in diminished bactericidal activity, thus protecting E. coli K1 from the elimination by neutrophils.
Collapse
Affiliation(s)
- Xue-Wei Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, China
| | - Ming-Xin An
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, China
| | - Zeng-Kang Huang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, China
| | - Lan Ma
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, China
| | - Dan Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, China,Department of Neurosurgery, the First Hospital of China Medical University, 155 Nanjing Street, Heping District, Shenyang, 110001, China
| | - Zhao Yang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, China
| | - Jun-Xiu Shi
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, China
| | - Dong-Xin Liu
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, China
| | - Qiang Li
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, 16 Puhe Road, Shenbei New District, Shenyang, 110134, China
| | - An-Hua Wu
- Department of Neurosurgery, the First Hospital of China Medical University, 155 Nanjing Street, Heping District, Shenyang, 110001, China
| | - Yu-Hua Chen
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, China
| | - Wei-Dong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, China.
| |
Collapse
|
44
|
Yang E, Sekhon M, Griesdale D. Transtentorial herniation syndrome from meningococcal meningitis in a young woman: the case for neurocritical care. BMJ Case Rep 2022; 15:e253191. [PMID: 36450413 PMCID: PMC9716947 DOI: 10.1136/bcr-2022-253191] [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] [Accepted: 11/21/2022] [Indexed: 12/05/2022] Open
Abstract
We report a case of a previously healthy early adolescent female who presented with meningococcal meningitis. While in hospital, she had marked neurologic deterioration with clinical herniation from malignant cerebral oedema. She was transferred to a neurocritical care centre where she underwent invasive intracranial pressure (ICP) and brain tissue oxygen (PbtO2) monitoring. Early in her course, she demonstrated a compete absence of autoregulation, with pressure passive cerebral blood flow. As a result, maintaining a mean arterial pressure between 50 mm Hg and 60 mm Hg, which ensured adequate cerebral oxygenation, while avoiding increases in ICP. Although her course was initially complicated by bilateral optic neuropathy, she has subsequently made a full neurologic recovery and is now undertaking postsecondary education. This case highlights that access to specialist neurocritical care, guided by neurophysiologic monitoring of ICP and PbtO2, may help improve outcomes, even among those patients with catastrophic cerebral oedema from bacterial meningitis.
Collapse
Affiliation(s)
- Eleen Yang
- Department of Anesthesiology, Pharmacology & Therapeutics, The University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada
| | - Mypinder Sekhon
- Department of Medicine, Division of Critical Care Medicine, The University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada
| | - Donald Griesdale
- Department of Anesthesiology, Pharmacology & Therapeutics, The University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada
| |
Collapse
|
45
|
Tan MF, Tan J, Zhang FF, Li HQ, Ji HY, Fang SP, Wu CC, Rao YL, Zeng YB, Yang Q. Exogenous glycogen utilization effects the transcriptome and pathogenicity of Streptococcus suis serotype 2. Front Cell Infect Microbiol 2022; 12:938286. [PMID: 36439226 PMCID: PMC9683343 DOI: 10.3389/fcimb.2022.938286] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 10/24/2022] [Indexed: 09/23/2024] Open
Abstract
Streptococcus suis serotype 2 (SS2) is an important zoonotic pathogen that causes severe infections in humans and the swine industry. Acquisition and utilization of available carbon sources from challenging host environments is necessary for bacterial pathogens to ensure growth and proliferation. Glycogen is abundant in mammalian body and may support the growth of SS2 during infection in hosts. However, limited information is known about the mechanism between the glycogen utilization and host adaptation of SS2. Here, the pleiotropic effects of exogenous glycogen on SS2 were investigated through transcriptome sequencing. Analysis of transcriptome data showed that the main basic metabolic pathways, especially the core carbon metabolism pathways and virulence-associated factors, of SS2 responded actively to glycogen induction. Glycogen induction led to the perturbation of the glycolysis pathway and citrate cycle, but promoted the pentose phosphate pathway and carbohydrate transport systems. Extracellular glycogen utilization also promoted the mixed-acid fermentation in SS2 rather than homolactic fermentation. Subsequently, apuA, a gene encoding the unique bifunctional amylopullulanase for glycogen degradation, was deleted from the wild type and generated the mutant strain ΔapuA. The pathogenicity details of the wild type and ΔapuA cultured in glucose and glycogen were investigated and compared. Results revealed that the capsule synthesis or bacterial morphology were not affected by glycogen incubation or apuA deletion. However, extracellular glycogen utilization significantly enhanced the hemolytic activity, adhesion and invasion ability, and lethality of SS2. The deletion of apuA also impaired the pathogenicity of bacteria cultured in glucose, indicating that ApuA is indeed an important virulence factor. Our results revealed that exogenous glycogen utilization extensively influenced the expression profile of the S. suis genome. Based on the transcriptome response, exogenous glycogen utilization promoted the carbon adaption and pathogenicity of SS2.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yan-Bin Zeng
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang, China
| | - Qun Yang
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang, China
| |
Collapse
|
46
|
Endres LM, Jungblut M, Divyapicigil M, Sauer M, Stigloher C, Christodoulides M, Kim BJ, Schubert-Unkmeir A. Development of a multicellular in vitro model of the meningeal blood-CSF barrier to study Neisseria meningitidis infection. Fluids Barriers CNS 2022; 19:81. [PMID: 36289516 PMCID: PMC9597984 DOI: 10.1186/s12987-022-00379-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/06/2022] [Indexed: 12/01/2022] Open
Abstract
Background Bacterial meningitis is a life-threatening disease that occurs when pathogens such as Neisseria meningitidis cross the meningeal blood cerebrospinal fluid barrier (mBCSFB) and infect the meninges. Due to the human-specific nature of N. meningitidis, previous research investigating this complex host–pathogen interaction has mostly been done in vitro using immortalized brain endothelial cells (BECs) alone, which often do not retain relevant barrier properties in culture. Here, we developed physiologically relevant mBCSFB models using BECs in co-culture with leptomeningeal cells (LMCs) to examine N. meningitidis interaction. Methods We used BEC-like cells derived from induced pluripotent stem cells (iBECs) or hCMEC/D3 cells in co-culture with LMCs derived from tumor biopsies. We employed TEM and structured illumination microscopy to characterize the models as well as bacterial interaction. We measured TEER and sodium fluorescein (NaF) permeability to determine barrier tightness and integrity. We then analyzed bacterial adherence and penetration of the cell barrier and examined changes in host gene expression of tight junctions as well as chemokines and cytokines in response to infection. Results Both cell types remained distinct in co-culture and iBECs showed characteristic expression of BEC markers including tight junction proteins and endothelial markers. iBEC barrier function as determined by TEER and NaF permeability was improved by LMC co-culture and remained stable for seven days. BEC response to N. meningitidis infection was not affected by LMC co-culture. We detected considerable amounts of BEC-adherent meningococci and a relatively small number of intracellular bacteria. Interestingly, we discovered bacteria traversing the BEC-LMC barrier within the first 24 h post-infection, when barrier integrity was still high, suggesting a transcellular route for N. meningitidis into the CNS. Finally, we observed deterioration of barrier properties including loss of TEER and reduced expression of cell-junction components at late time points of infection. Conclusions Here, we report, for the first time, on co-culture of human iPSC derived BECs or hCMEC/D3 with meningioma derived LMCs and find that LMC co-culture improves barrier properties of iBECs. These novel models allow for a better understanding of N. meningitidis interaction at the mBCSFB in a physiologically relevant setting. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00379-z.
Collapse
Affiliation(s)
- Leo M. Endres
- grid.8379.50000 0001 1958 8658Institute for Hygiene and Microbiology, University of Würzburg, Josef-Schneider-Strasse 2, 97080 Würzburg, Germany
| | - Marvin Jungblut
- grid.8379.50000 0001 1958 8658Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Mustafa Divyapicigil
- grid.411015.00000 0001 0727 7545Department of Biological Sciences, University of Alabama, Tuscaloosa, AL USA ,grid.265892.20000000106344187Department of Microbiology Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,grid.411015.00000 0001 0727 7545Center for Convergent Biosciences & Medicine, University of Alabama, Tuscaloosa, AL USA ,grid.411015.00000 0001 0727 7545Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL USA
| | - Markus Sauer
- grid.8379.50000 0001 1958 8658Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Christian Stigloher
- grid.8379.50000 0001 1958 8658Imaging Core Facility, Biocenter, University of Würzburg, Würzburg, Germany
| | - Myron Christodoulides
- grid.5491.90000 0004 1936 9297Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Brandon J. Kim
- grid.411015.00000 0001 0727 7545Department of Biological Sciences, University of Alabama, Tuscaloosa, AL USA ,grid.265892.20000000106344187Department of Microbiology Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,grid.411015.00000 0001 0727 7545Center for Convergent Biosciences & Medicine, University of Alabama, Tuscaloosa, AL USA ,grid.411015.00000 0001 0727 7545Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL USA
| | - Alexandra Schubert-Unkmeir
- grid.8379.50000 0001 1958 8658Institute for Hygiene and Microbiology, University of Würzburg, Josef-Schneider-Strasse 2, 97080 Würzburg, Germany
| |
Collapse
|
47
|
Bergmann S, Fulde M, Siemens N. Editorial: Streptococci in infectious diseases - pathogenic mechanisms and host immune responses. Front Microbiol 2022; 13:988671. [PMID: 36033862 PMCID: PMC9405650 DOI: 10.3389/fmicb.2022.988671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Simone Bergmann
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Marcus Fulde
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Free University of Berlin, Berlin, Germany
| | - Nikolai Siemens
- Department of Molecular Genetics and Infection Biology, University of Greifswald, Greifswald, Germany
| |
Collapse
|
48
|
Li YT, Li CX, Huang CJ, Wen QY, Deng SM, Zhu LP, Liu ZF, Chen ZG. Meconium-Stained Amniotic Fluid: Impact on Prognosis of Neonatal Bacterial Meningitis. J Trop Pediatr 2022; 68:6665809. [PMID: 35962981 DOI: 10.1093/tropej/fmac064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Clinical data with respect to the impact of meconium on the prognosis of neonatal bacterial meningitis are scarce. Therefore, in this study, we aimed to determine whether meconium-stained amniotic fluid (MSAF) represents a risk factor for poor prognosis of neonatal bacterial meningitis in a confirmed case population. METHODS This was a retrospective cohort study of 256 neonates diagnosed with bacterial meningitis hospitalized at one of three hospitals in Shantou, China, between October 2013 and September 2018. Clinical manifestation, laboratory test results and treatment were compared between the two groups, with outcomes dichotomized into 'good' or 'poor' prognosis. Multivariate analysis and follow-up logistic regression analysis were used to identify predictive factors of a poor outcome. RESULTS Of the 256 neonates with BM, 95 (37.1%) had a good prognosis at discharge and 161 (62.9%) had a poor prognosis. In the poor prognosis group, 131/161 (79.4%) neonates had a permanent neurological sequelae and 19 (11.8%) had ≥2 sequelae. Of note, 11 neonates died. The rate of poor prognosis of BM was significantly higher among neonates with than without MSAF (26.1% vs. 12.6%, respectively; p < 0.05). A logistic multivariate analysis to evaluate the prognostic effect of MSAF to BM showed that neonatal with MSAF is more likely to have a worse prognosis of BM [unadjusted odds ratio (OR), 2.44, 95% confidence interval (CI), 1.24-5.10; adjusted OR, 2.31; 95% CI, 1.09-5.17]. CONCLUSION MSAF is significantly associated with poor prognosis of neonatal bacterial meningitis. Therefore, in case of MSAF, more attention should be paid to neonatal bacterial meningitis.
Collapse
Affiliation(s)
- Ya-Ting Li
- Department of Pediatrics, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Cai-Xia Li
- Department of Pediatrics, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Chu-Jun Huang
- Department of Pediatrics, Shantou Central Hospital, Shantou, China
| | - Qian-Yu Wen
- Clinical Data Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shu-Min Deng
- Department of Pediatrics, Shantou Central Hospital, Shantou, China
| | - Ling-Ping Zhu
- Department of Pediatrics, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zi-Feng Liu
- Clinical Data Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhuang-Gui Chen
- Department of Pediatrics, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
49
|
SssP1, a Fimbria-like component of Streptococcus suis, binds to the vimentin of host cells and contributes to bacterial meningitis. PLoS Pathog 2022; 18:e1010710. [PMID: 35853077 PMCID: PMC9337661 DOI: 10.1371/journal.ppat.1010710] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 07/29/2022] [Accepted: 06/27/2022] [Indexed: 11/19/2022] Open
Abstract
Streptococcus suis (S. suis) is one of the important pathogens that cause bacterial meningitis in pigs and humans. Evading host immune defences and penetrating the blood-brain barrier (BBB) are the preconditions for S. suis to cause meningitis, while the underlying mechanisms during these pathogenic processes are not fully understood. By detecting the red blood and white blood cells counts, IL-8 expression, and the pathological injury of brain in a mouse infection model, a serine-rich repeat (SRR) glycoprotein, designated as SssP1, was identified as a critical facilitator in the process of causing meningitis in this study. SssP1 was exported to assemble a fimbria-like component, thus contributed to the bacterial adhesion to and invasion into human brain microvascular endothelial cells (HBMECs), and activates the host inflammatory response during meningitis but is not involved in the actin cytoskeleton rearrangement and the disruption of tight junctions. Furthermore, the deletion of sssP1 significantly attenuates the ability of S. suis to traverse the BBB in vivo and in vitro. A pull-down analysis identified vimentin as the potential receptors of SssP1 during meningitis and following Far-Western blot results confirmed this ligand-receptor binding mediated by the NR2 (the second nonrepeat region) region of SssP1. The co-localisation of vimentin and S. suis observed by laser scanning confocal microscopy with multiplex fluorescence indicated that vimentin significantly enhances the interaction between SssP1 and BBB. Further study identified that the NR216-781 and NR1711-2214 fragments of SssP1 play critical roles to bind to the BBB depending on the sialylation of vimentin, and this binding is significantly attenuated when the antiserum of NR216-781 or NR1711-2214 blocked the bacterial cells, or the vimentin antibody blocked the BBB. Similar binding attenuations are observed when the bacterial cells were preincubated with the vimentin, or the BBB was preincubated with the recombinant protein NR216-781, NR1711-2214 or sialidase. In conclusion, these results reveal a novel receptor-ligand interaction that enhances adhesion to and penetration of the BBB to cause bacterial meningitis in the S. suis infection and highlight the importance of vimentin in host-pathogen interactions. Streptococcus suis (S. suis) is considered an important zoonotic pathogen capable of causing meningitis in humans. Penetrating the blood-brain barrier (BBB) is one of the preconditions for S. suis to cause meningitis, while its underlying mechanism is incompletely understood. Here we identified a previously uncharacterised pathogenic mechanism associated with S. suis meningitis mediated by the interaction between bacterial SRR glycoproteins and a host cytoskeletal component. During the bacterial infection, SRR protein SssP1 is exported to assemble a fimbria-like component, which drives a strong binding effect with the BBB depending on the sialylation of vimentin. This interaction contributes to the bacterial adhesion to and penetration of the BBB and induces a robust inflammatory response during meningitis. This overall observation underscores the significance of host cell surface vimentin interactions in microbial pathogenesis and markedly improves our understanding of host barrier penetration during meningitis.
Collapse
|
50
|
Sui Y, Chen Y, Lv Q, Zheng Y, Kong D, Jiang H, Huang W, Ren Y, Liu P, Jiang Y. Suilyin Disrupts the Blood-Brain Barrier by Activating Group III Secretory Phospholipase A2. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060919. [PMID: 35743951 PMCID: PMC9229629 DOI: 10.3390/life12060919] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/25/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022]
Abstract
Serious diseases caused by Streptococcus suis serotype 2 (S. suis 2) include septicaemia and meningitis, which are associated with high morbidity and mortality. Proliferation in the blood can result in a breach of the blood-brain barrier (BBB) and provide entry into the cerebrospinal fluid (CSF), where bacteria cause inflammation of the meningeal membranes resulting in meningitis. The molecular mechanisms of how this pathogen crosses the BBB remain unclear. Suilysin (SLY) has been identified as an important secreted virulence factor of S. suis 2 and may play a vital role in provoking meningitis. In this investigation, we demonstrate that SLY can increase the paracellular permeability of BBB, both in vivo and in vitro, via the activation of group III secretory phospholipase A2 (PLA2G3). Our results indicate that at lower, sublytic concentrations, the toxin can stimulate cerebral microvascular endothelial cells to release TNF-α, thereby inducing high level expressions of PLA2G3. Abnormal elevations of PLA2G3 might further injure tissues through direct cytolytic effectors or other responses.
Collapse
Affiliation(s)
- Yutong Sui
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing 100071, China; (Y.S.); (Q.L.); (Y.Z.); (D.K.); (H.J.); (W.H.); (Y.R.)
| | - Ying Chen
- School of Light Industry, Beijing Technology and Business University (BTBU), Beijing 100048, China;
| | - Qingyu Lv
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing 100071, China; (Y.S.); (Q.L.); (Y.Z.); (D.K.); (H.J.); (W.H.); (Y.R.)
| | - Yuling Zheng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing 100071, China; (Y.S.); (Q.L.); (Y.Z.); (D.K.); (H.J.); (W.H.); (Y.R.)
| | - Decong Kong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing 100071, China; (Y.S.); (Q.L.); (Y.Z.); (D.K.); (H.J.); (W.H.); (Y.R.)
| | - Hua Jiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing 100071, China; (Y.S.); (Q.L.); (Y.Z.); (D.K.); (H.J.); (W.H.); (Y.R.)
| | - Wenhua Huang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing 100071, China; (Y.S.); (Q.L.); (Y.Z.); (D.K.); (H.J.); (W.H.); (Y.R.)
| | - Yuhao Ren
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing 100071, China; (Y.S.); (Q.L.); (Y.Z.); (D.K.); (H.J.); (W.H.); (Y.R.)
| | - Peng Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing 100071, China; (Y.S.); (Q.L.); (Y.Z.); (D.K.); (H.J.); (W.H.); (Y.R.)
- Correspondence: (P.L.); (Y.J.)
| | - Yongqiang Jiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing 100071, China; (Y.S.); (Q.L.); (Y.Z.); (D.K.); (H.J.); (W.H.); (Y.R.)
- Correspondence: (P.L.); (Y.J.)
| |
Collapse
|