1
|
García E. Structure, Function, and Regulation of LytA: The N-Acetylmuramoyl-l-alanine Amidase Driving the "Suicidal Tendencies" of Streptococcus pneumoniae-A Review. Microorganisms 2025; 13:827. [PMID: 40284663 PMCID: PMC12029793 DOI: 10.3390/microorganisms13040827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
Streptococcus pneumoniae (pneumococcus) is a significant human pathogen responsible for a range of diseases from mild infections to invasive pneumococcal diseases, particularly affecting children, the elderly, and immunocompromised individuals. Despite pneumococcal conjugate vaccines having reduced disease incidence, challenges persist due to serotype diversity, vaccine coverage gaps, and antibiotic resistance. This review highlights the role of LytA, a key autolysin (N-acetylmuramoyl-l-alanine amidase), in pneumococcal biology. LytA regulates autolysis, contributes to inflammation, and biofilm formation, and impairs bacterial clearance. It also modulates complement activation, aiding immune evasion. LytA expression is influenced by environmental signals and genetic regulation and is tied to competence for genetic transformation, which is an important virulence trait, particularly in meningitis. With the increase in antibiotic resistance, LytA has emerged as a potential therapeutic target. Current research explores its use in bacteriolytic therapies, vaccine development, and synergistic antibiotic strategies. Various compounds, including synthetic peptides, plant extracts, and small molecules, have been investigated for their ability to trigger LytA-mediated bacterial lysis. Future directions include the development of novel anti-pneumococcal interventions leveraging LytA's properties while overcoming vaccine efficacy and resistance-related challenges. Human challenge models and animal studies continue to deepen our understanding of pneumococcal pathogenesis and potential treatment strategies.
Collapse
Affiliation(s)
- Ernesto García
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, 28040 Madrid, Spain
| |
Collapse
|
2
|
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
|
3
|
Balde A, Ramya CS, Nazeer RA. A review on current advancement in zebrafish models to study chronic inflammatory diseases and their therapeutic targets. Heliyon 2024; 10:e31862. [PMID: 38867970 PMCID: PMC11167310 DOI: 10.1016/j.heliyon.2024.e31862] [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: 11/15/2023] [Revised: 04/02/2024] [Accepted: 05/22/2024] [Indexed: 06/14/2024] Open
Abstract
Chronic inflammatory diseases are caused due to prolonged inflammation at a specific site of the body. Among other inflammatory diseases, bacterial meningitis, chronic obstructive pulmonary disease (COPD), atherosclerosis and inflammatory bowel diseases (IBD) are primarily focused on because of their adverse effects and fatality rates around the globe in recent times. In order to come up with novel strategies to eradicate these diseases, a clear understanding of the mechanisms of the diseases is needed. Similarly, detailed insight into the mechanisms of commercially available drugs and potent lead compounds from natural sources are also important to establish efficient therapeutic effects. Zebrafish is widely accepted as a model to study drug toxicity and the pharmacokinetic effects of the drug. Moreover, researchers use various inducers to trigger inflammatory cascades and stimulate physiological changes in zebrafish. The effect of these inducers contrasts with the type of zebrafish used in the investigation. Hence, a thorough analysis is required to study the current advancements in the zebrafish model for chronic inflammatory disease suppression. This review presents the most common inflammatory diseases, commercially available drugs, novel therapeutics, and their mechanisms of action for disease suppression. The review also provides a detailed description of various zebrafish models for these diseases. Finally, the future prospects and challenges for the same are described, which can help the researchers understand the potency of the zebrafish model and its further exploration for disease attenuation.
Collapse
Affiliation(s)
- Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Cunnathur Saravanan Ramya
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| |
Collapse
|
4
|
Shen Z, Ke Z, Yang Q, Ghebremichael ST, Li T, Li T, Chen J, Meng X, Xiang H, Li C, Zhou Z, Pan G, Chen P. Transcriptomic changes in the microsporidia proliferation and host responses in congenitally infected embryos and larvae. BMC Genomics 2024; 25:321. [PMID: 38556880 PMCID: PMC10983672 DOI: 10.1186/s12864-024-10236-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 03/18/2024] [Indexed: 04/02/2024] Open
Abstract
Congenital infection caused by vertical transmission of microsporidia N. bombycis can result in severe economic losses in the silkworm-rearing industry. Whole-transcriptome analyses have revealed non-coding RNAs and their regulatory networks in N. bombycis infected embryos and larvae. However, transcriptomic changes in the microsporidia proliferation and host responses in congenitally infected embryos and larvae remains unclear. Here, we simultaneously compared the transcriptomes of N. bombycis and its host B. mori embryos of 5-day and larvae of 1-, 5- and 10-day during congenital infection. For the transcriptome of N. bombycis, a comparison of parasite expression patterns between congenital-infected embryos and larva showed most genes related to parasite central carbon metabolism were down-regulated in larvae during infection, whereas the majority of genes involved in parasite proliferation and growth were up-regulated. Interestingly, a large number of distinct or shared differentially expressed genes (DEGs) were revealed by the Venn diagram and heat map, many of them were connected to infection related factors such as Ricin B lectin, spore wall protein, polar tube protein, and polysaccharide deacetylase. For the transcriptome of B. mori infected with N. bombycis, beyond numerous DEGs related to DNA replication and repair, mRNA surveillance pathway, RNA transport, protein biosynthesis, and proteolysis, with the progression of infection, a large number of DEGs related to immune and infection pathways, including phagocytosis, apoptosis, TNF, Toll-like receptor, NF-kappa B, Fc epsilon RI, and some diseases, were successively identified. In contrast, most genes associated with the insulin signaling pathway, 2-oxacarboxylic acid metabolism, amino acid biosynthesis, and lipid metabolisms were up-regulated in larvae compared to those in embryos. Furthermore, dozens of distinct and three shared DEGs that were involved in the epigenetic regulations, such as polycomb, histone-lysine-specific demethylases, and histone-lysine-N-methyltransferases, were identified via the Venn diagram and heat maps. Notably, many DEGs of host and parasite associated with lipid-related metabolisms were verified by RT-qPCR. Taken together, simultaneous transcriptomic analyses of both host and parasite genes lead to a better understanding of changes in the microsporidia proliferation and host responses in embryos and larvae in N. bombycis congenital infection.
Collapse
Affiliation(s)
- Zigang Shen
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Tiansheng Street, Chongqing, 400716, China
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Zhuojun Ke
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Qiong Yang
- Sericulture and Agri-food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Samson Teweldeberhan Ghebremichael
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Tangxin Li
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Tian Li
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Jie Chen
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Xianzhi Meng
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Heng Xiang
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Chunfeng Li
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
| | - Zeyang Zhou
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China
- College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Guoqing Pan
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China.
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Tiansheng Street, Chongqing, 400716, China.
| | - Ping Chen
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Tiansheng Street, Chongqing, 400716, China.
- State Key Laboratory of Resource Insects, Southwest University, Tiansheng Street, Chongqing, 400716, China.
| |
Collapse
|
5
|
Patel P, Nandi A, Verma SK, Kaushik N, Suar M, Choi EH, Kaushik NK. Zebrafish-based platform for emerging bio-contaminants and virus inactivation research. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 872:162197. [PMID: 36781138 PMCID: PMC9922160 DOI: 10.1016/j.scitotenv.2023.162197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/23/2023] [Accepted: 02/08/2023] [Indexed: 05/27/2023]
Abstract
Emerging bio-contaminants such as viruses have affected health and environment settings of every country. Viruses are the minuscule entities resulting in severe contagious diseases like SARS, MERS, Ebola, and avian influenza. Recent epidemic like the SARS-CoV-2, the virus has undergone mutations strengthen them and allowing to escape from the remedies. Comprehensive knowledge of viruses is essential for the development of targeted therapeutic and vaccination treatments. Animal models mimicking human biology like non-human primates, rats, mice, and rabbits offer competitive advantage to assess risk of viral infections, chemical toxins, nanoparticles, and microbes. However, their economic maintenance has always been an issue. Furthermore, the redundancy of experimental results due to aforementioned aspects is also in examine. Hence, exploration for the alternative animal models is crucial for risk assessments. The current review examines zebrafish traits and explores the possibilities to monitor emerging bio-contaminants. Additionally, a comprehensive picture of the bio contaminant and virus particle invasion and abatement mechanisms in zebrafish and human cells is presented. Moreover, a zebrafish model to investigate the emerging viruses such as coronaviridae and poxviridae has been suggested.
Collapse
Affiliation(s)
- Paritosh Patel
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea
| | - Aditya Nandi
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Suresh K Verma
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India; Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, 18323 Hwaseong, Republic of Korea
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| |
Collapse
|
6
|
Saralahti AK, Harjula SKE, Rantapero T, Uusi-Mäkelä MIE, Kaasinen M, Junno M, Piippo H, Nykter M, Lohi O, Rounioja S, Parikka M, Rämet M. Characterization of the innate immune response to Streptococcus pneumoniae infection in zebrafish. PLoS Genet 2023; 19:e1010586. [PMID: 36622851 PMCID: PMC9858863 DOI: 10.1371/journal.pgen.1010586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 01/20/2023] [Accepted: 12/20/2022] [Indexed: 01/10/2023] Open
Abstract
Streptococcus pneumoniae (pneumococcus) is one of the most frequent causes of pneumonia, sepsis and meningitis in humans, and an important cause of mortality among children and the elderly. We have previously reported the suitability of the zebrafish (Danio rerio) larval model for the study of the host-pathogen interactions in pneumococcal infection. In the present study, we characterized the zebrafish innate immune response to pneumococcus in detail through a whole-genome level transcriptome analysis and revealed a well-conserved response to this human pathogen in challenged larvae. In addition, to gain understanding of the genetic factors associated with the increased risk for severe pneumococcal infection in humans, we carried out a medium-scale forward genetic screen in zebrafish. In the screen, we identified a mutant fish line which showed compromised resistance to pneumococcus in the septic larval infection model. The transcriptome analysis of the mutant zebrafish larvae revealed deficient expression of a gene homologous for human C-reactive protein (CRP). Furthermore, knockout of one of the six zebrafish crp genes by CRISPR-Cas9 mutagenesis predisposed zebrafish larvae to a more severe pneumococcal infection, and the phenotype was further augmented by concomitant knockdown of a gene for another Crp isoform. This suggests a conserved function of C-reactive protein in anti-pneumococcal immunity in zebrafish. Altogether, this study highlights the similarity of the host response to pneumococcus in zebrafish and humans, gives evidence of the conserved role of C-reactive protein in the defense against pneumococcus, and suggests novel host genes associated with pneumococcal infection.
Collapse
Affiliation(s)
- Anni K. Saralahti
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sanna-Kaisa E. Harjula
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Tommi Rantapero
- Laboratory of Computational Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Meri I. E. Uusi-Mäkelä
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mikko Kaasinen
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Maiju Junno
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Hannaleena Piippo
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Matti Nykter
- Laboratory of Computational Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Olli Lohi
- Tampere Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | | | - Mataleena Parikka
- Laboratory of Infection Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mika Rämet
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- FVR–Finnish Vaccine Research, Tampere, Finland
| |
Collapse
|
7
|
Jacome Sanz D, Saralahti AK, Pekkarinen M, Kesseli J, Nykter M, Rämet M, Ojanen MJT, Pesu M. Proprotein convertase subtilisin/kexin type 9 regulates the production of acute-phase reactants from the liver. Liver Int 2021; 41:2511-2522. [PMID: 34174143 DOI: 10.1111/liv.14993] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/07/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Proprotein convertase subtilisin/kexin type 9 (PCSK9) controls blood cholesterol levels by fostering the LDL receptor (LDLR) degradation in hepatocytes. Additionally, PCSK9 has been suggested to participate in immunoregulation by modulating cytokine production. We studied the immunological role of PCSK9 in Streptococcus pneumoniae bacteraemia in vivo and in a human hepatocyte cell line. METHODS CRISPR/Cas9 mutagenesis was utilized to create pcsk9 knock-out (KO) zebrafish, which were infected with S pneumoniae to assess the role of PCSK9 for the survival of the fish and in the transcriptomic response of the liver. The direct effects of PCSK9 on the expression of acute-phase reaction (APR) genes were studied in HepG2 cells. RESULTS The pcsk9 KO zebrafish lines (pcsk9tpu-13 and pcsk9tpu-2,+15 ) did not show developmental defects or gross phenotypical differences. In the S pneumoniae infected zebrafish, the mortality of pcsk9 KOs was similar to the controls. A liver-specific gene expression analysis revealed that a pneumococcal challenge upregulated pcsk9, and that the pcsk9 deletion reduced the expression of APR genes, including hepcidin antimicrobial peptide (hamp) and complement component 7b (c7b). Accordingly, silencing PCSK9 in vitro in HepG2 cells using small interfering RNAs (siRNAs) decreased HAMP expression. CONCLUSIONS We demonstrate that PCSK9 is not critical for zebrafish survival in a systemic pneumococcal infection. However, PCSK9 deficiency was associated with the lower expression of APR genes in zebrafish and altered the expression of innate immunity genes in a human hepatocyte cell line. Overall, our data suggest an evolutionarily conserved function for PCSK9 in APR in the liver.
Collapse
Affiliation(s)
- Dafne Jacome Sanz
- Laboratory of Immunoregulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Anni K Saralahti
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Meeri Pekkarinen
- Laboratory of Computational Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Juha Kesseli
- Laboratory of Computational Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Matti Nykter
- Laboratory of Computational Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mika Rämet
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,PEDEGO Research Unit, Medical Research Center, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Markus J T Ojanen
- Laboratory of Immunoregulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Marko Pesu
- Laboratory of Immunoregulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Fimlab laboratories Ltd, Tampere, Finland
| |
Collapse
|
8
|
Sharma S, Kumar M, Kumar J, Srivastava N, Hussain MA, Shelly A, Mazumder S. M. fortuitum-induced CNS-pathology: Deciphering the role of canonical Wnt signaling, blood brain barrier components and cytokines. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 122:104111. [PMID: 33933535 DOI: 10.1016/j.dci.2021.104111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/24/2021] [Accepted: 04/24/2021] [Indexed: 06/12/2023]
Abstract
Molecular underpinning of mycobacteria-induced CNS-pathology is not well understood. In the present study, zebrafish were infected with Mycobacterium fortuitum and the prognosis of CNS-pathogenesis studied. We observed M. fortuitum triggers extensive brain-pathology. Evans blue extravasation demonstrated compromised blood-brain barrier (BBB) integrity. Further, decreased expression in tight-junction (TJ) and adherens junction complex (AJC) genes were noted in infected brain. Wnt-signaling has emerged as a major player in host-mycobacterial immunity but its involvement/role in brain-infection is not well studied. Sustained expression of wnt2, wnt3a, fzd5, lrp5/6 and β-catenin, with concordant decline in degradation complex components axin, gsk3β and β-catenin regulator capn2a were observed. The surge in ifng1 and tnfa expression preceding il10 and il4 suggested cytokine-interplay critical in M. fortuitum-induced brain-pathology. Therefore, we suggest adult zebrafish as a viable model for studying CNS-pathology and using the same, conclude that M. fortuitum infection is associated with repressed TJ-AJC gene expression and compromised BBB permeability. Our results implicate Wnt/β-catenin pathway in M. fortuitum-induced CNS-pathology wherein Th1-type signals facilitate bacterial clearance and Th2-type signals prevent the disease sequel.
Collapse
Affiliation(s)
- Shagun Sharma
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Manmohan Kumar
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Jai Kumar
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Nidhi Srivastava
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India; Department of Zoology, School of Basic and Applied Sciences, Maharaja Agrasen University, Solan, Himachal Pradesh, 174103, India
| | - Md Arafat Hussain
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Asha Shelly
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Shibnath Mazumder
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India; Faculty of Life Sciences and Biotechnology, South Asian University, Delhi, 110021, India.
| |
Collapse
|
9
|
Lin JN, Lai CH, Huang YH, Yang CH. Antimicrobial Effects of Minocycline, Tigecycline, Ciprofloxacin, and Levofloxacin against Elizabethkingia anophelis Using In Vitro Time-Kill Assays and In Vivo Zebrafish Animal Models. Antibiotics (Basel) 2021; 10:antibiotics10030285. [PMID: 33801839 PMCID: PMC7999888 DOI: 10.3390/antibiotics10030285] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/19/2022] Open
Abstract
Elizabethkingia anophelis is a multidrug-resistant pathogen. This study evaluated the antimicrobial activity of minocycline, tigecycline, ciprofloxacin, and levofloxacin using in vitro time-kill assays and in vivo zebrafish animal models. The E. anophelis strain ED853-49 was arbitrarily selected from a bacterial collection which was concomitantly susceptible to minocycline, tigecycline, ciprofloxacin, and levofloxacin. The antibacterial activities of single agents at 0.5-4 × minimum inhibitory concentration (MIC) and dual-agent combinations at 2 × MIC using time-kill assays were investigated. The therapeutic effects of antibiotics in E. anophelis-infected zebrafish were examined. Both minocycline and tigecycline demonstrated bacteriostatic effects but no bactericidal effect. Minocycline at concentrations ≥2 × MIC and tigecycline at concentrations ≥3 × MIC exhibited a long-standing inhibitory effect for 48 h. Bactericidal effects were observed at ciprofloxacin and levofloxacin concentrations of ≥3 × MIC within 24 h of initial inoculation. Rapid regrowth of E. anophelis occurred after the initial killing phase when ciprofloxacin was used, regardless of the concentration. Levofloxacin treatment at the concentration of ≥2 × MIC consistently resulted in the long-lasting and sustainable inhibition of bacterial growth for 48 h. The addition of minocycline or tigecycline weakened the killing effect of fluoroquinolones during the first 10 h. The minocycline-ciprofloxacin or minocycline-levofloxacin combinations achieved the lowest colony-forming unit counts at 48 h. Zebrafish treated with minocycline or a combination of minocycline and levofloxacin had the highest survival rate (70%). The results of these in vitro and in vivo studies suggest that the combination of minocycline and levofloxacin is the most effective therapy approach for E. anophelis infection.
Collapse
Affiliation(s)
- Jiun-Nong Lin
- Department of Critical Care Medicine, E-Da Hospital, I-Shou University, Kaohsiung 824, Taiwan
- Division of Infectious Diseases, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung 824, Taiwan;
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 824, Taiwan;
- Correspondence:
| | - Chung-Hsu Lai
- Division of Infectious Diseases, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung 824, Taiwan;
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 824, Taiwan;
| | - Yi-Han Huang
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 824, Taiwan;
| | - Chih-Hui Yang
- Department of Biological Science and Technology, Meiho University, Pingtung 912, Taiwan;
| |
Collapse
|
10
|
Liu X, Kimmey JM, Matarazzo L, de Bakker V, Van Maele L, Sirard JC, Nizet V, Veening JW. Exploration of Bacterial Bottlenecks and Streptococcus pneumoniae Pathogenesis by CRISPRi-Seq. Cell Host Microbe 2020; 29:107-120.e6. [PMID: 33120116 DOI: 10.1016/j.chom.2020.10.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 08/27/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023]
Abstract
Streptococcus pneumoniae is an opportunistic human pathogen that causes invasive diseases, including pneumonia, with greater health risks upon influenza A virus (IAV) co-infection. To facilitate pathogenesis studies in vivo, we developed an inducible CRISPR interference system that enables genome-wide fitness testing in one sequencing step (CRISPRi-seq). We applied CRISPRi-seq to assess bottlenecks and identify pneumococcal genes important in a murine pneumonia model. A critical bottleneck occurs at 48 h with few bacteria causing systemic infection. This bottleneck is not present during IAV superinfection, facilitating identification of pneumococcal pathogenesis-related genes. Top in vivo essential genes included purA, encoding adenylsuccinate synthetase, and the cps operon required for capsule production. Surprisingly, CRISPRi-seq indicated no fitness-related role for pneumolysin during superinfection. Interestingly, although metK (encoding S-adenosylmethionine synthetase) was essential in vitro, it was dispensable in vivo. This highlights advantages of CRISPRi-seq over transposon-based genetic screens, as all genes, including essential genes, can be tested for pathogenesis potential.
Collapse
Affiliation(s)
- Xue Liu
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, Lausanne 1015, Switzerland
| | - Jacqueline M Kimmey
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Laura Matarazzo
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Vincent de Bakker
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, Lausanne 1015, Switzerland
| | - Laurye Van Maele
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Jean-Claude Sirard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Victor Nizet
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Jan-Willem Veening
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, Lausanne 1015, Switzerland.
| |
Collapse
|
11
|
Subramanian K, Iovino F, Tsikourkitoudi V, Merkl P, Ahmed S, Berry SB, Aschtgen MS, Svensson M, Bergman P, Sotiriou GA, Henriques-Normark B. Mannose receptor-derived peptides neutralize pore-forming toxins and reduce inflammation and development of pneumococcal disease. EMBO Mol Med 2020; 12:e12695. [PMID: 32985105 PMCID: PMC7645366 DOI: 10.15252/emmm.202012695] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/30/2020] [Accepted: 09/02/2020] [Indexed: 12/30/2022] Open
Abstract
Cholesterol‐dependent cytolysins (CDCs) are essential virulence factors for many human pathogens like Streptococcus pneumoniae (pneumolysin, PLY), Streptococcus pyogenes (streptolysin O, SLO), and Listeria monocytogenes (Listeriolysin, LLO) and induce cytolysis and inflammation. Recently, we identified that pneumococcal PLY interacts with the mannose receptor (MRC‐1) on specific immune cells thereby evoking an anti‐inflammatory response at sublytic doses. Here, we identified the interaction sites between MRC‐1 and CDCs using computational docking. We designed peptides from the CTLD4 domain of MRC‐1 that binds to PLY, SLO, and LLO, respectively. In vitro, the peptides blocked CDC‐induced cytolysis and inflammatory cytokine production by human macrophages. Also, they reduced PLY‐induced damage of the epithelial barrier integrity as well as blocked bacterial invasion into the epithelium in a 3D lung tissue model. Pre‐treatment of human DCs with peptides blocked bacterial uptake via MRC‐1 and reduced intracellular bacterial survival by targeting bacteria to autophagosomes. In order to use the peptides for treatment in vivo, we developed calcium phosphate nanoparticles (CaP NPs) as peptide nanocarriers for intranasal delivery of peptides and enhanced bioactivity. Co‐administration of peptide‐loaded CaP NPs during infection improved survival and bacterial clearance in both zebrafish and mice models of pneumococcal infection. We suggest that MRC‐1 peptides can be employed as adjunctive therapeutics with antibiotics to treat bacterial infections by countering the action of CDCs.
Collapse
Affiliation(s)
- Karthik Subramanian
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Federico Iovino
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Vasiliki Tsikourkitoudi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Padryk Merkl
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sultan Ahmed
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Samuel B Berry
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | | | - Mattias Svensson
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Peter Bergman
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden.,The Immunodeficiency Unit, Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Georgios A Sotiriou
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Birgitta Henriques-Normark
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden.,Lee Kong Chian School of Medicine (LKC) and Singapore Centre on Environmental Life Sciences Engineering (SCELSE), Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
12
|
Wu XM, Cao L, Hu YW, Chang MX. Transcriptomic characterization of adult zebrafish infected with Streptococcus agalactiae. FISH & SHELLFISH IMMUNOLOGY 2019; 94:355-372. [PMID: 31533079 DOI: 10.1016/j.fsi.2019.09.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/12/2019] [Accepted: 09/15/2019] [Indexed: 06/10/2023]
Abstract
Streptococcus agalactiae is a major aquaculture pathogen infecting various saltwater and freshwater fish. To better understand the mechanism of the immune responses to S. agalactiae in wildtype zebrafish, the transcriptomic profiles of two organs containing mucosal-associated lymphoid tissues from S. agalactiae-infected and non-infected groups were obtained using RNA-seq techniques. In the intestines, 6735 and 12908 differently expressed genes (DEGs) were identified at 24 hpi and 48 hpi, respectively. Among 66 and 116 significantly enriched pathways, 15 and 21 pathways were involved in immune system or signal transduction at 24 hpi and 48 hpi, respectively. A number of genes involved in Toll-like receptor signaling pathway, RIG-I-like receptor signaling pathway, NOD-like receptor signaling pathway, T cell receptor signaling pathway, B cell receptor signaling pathway, Antigen processing and presentation, NF-kappa B signaling pathway and PI3K-Akt signaling pathway were significantly downregulated. In the skins, 3113 and 4467 DEGs were identified at 24 hpi and 48 hpi, respectively. Among 24 and 56 significantly enriched pathways, 4 and 13 pathways were involved in immune system or signal transduction at 24 hpi and 48 hpi, respectively. More immune-related signaling pathways including Leukocyte transendothelial migration, Cytokine-cytokine receptor interaction, PI3K-Akt signaling pathway, IL-17 signaling pathway, MAPK signaling pathway, TNF signaling pathway, Complement and coagulation cascades, Hematopoietic cell lineage and Jak-STAT signaling pathway were differently enriched for upregulated DEGs at 48 hpi, which were completely different from that in the intestines. Furthmore, comparative transcriptome analysis revealed that the downregulated 1618 genes and upregulated 1622 genes existed both at 24 hpi and 48 hpi for the intestine samples. In the skins, the downregulated 672 genes and upregulated 428 genes existed both at 24 hpi and 48 hpi. Three pathways related to immune processes were significantly enriched for downregulated DEGs both in the intestines and skins collected at 24 hpi and 48 hpi, which included Antigen processing and presentation, Intestinal immune network for IgA production and Hematopoietic cell lineage. Interaction network analysis of DEGs identified the main DEGs in the sub-network of complement and coagulation cascades both in the intestines and skins. Twenty of DEGs involved in complement and coagulation cascades were further validated by Real-time quantitative PCR. Altogether, the results obtained in this study will provide insight into the immune response of zebrafish against S. agalactiae XQ-1 infection in fatal conditions, and reveal the discrepant expression pattern of complement and coagulation cascades in the intestines and skins.
Collapse
Affiliation(s)
- Xiao Man Wu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, China; University of Chinese Academy of Sciences, Beijing, China
| | - Lu Cao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yi Wei Hu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ming Xian Chang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, China; Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan, Hubei Province, China; The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
13
|
Jagau H, Behrens IK, Lahme K, Lorz G, Köster RW, Schneppenheim R, Obser T, Brehm MA, König G, Kohler TP, Rohde M, Frank R, Tegge W, Fulde M, Hammerschmidt S, Steinert M, Bergmann S. Von Willebrand Factor Mediates Pneumococcal Aggregation and Adhesion in Blood Flow. Front Microbiol 2019; 10:511. [PMID: 30972039 PMCID: PMC6443961 DOI: 10.3389/fmicb.2019.00511] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/27/2019] [Indexed: 12/24/2022] Open
Abstract
Streptococcus pneumoniae is a major cause of community acquired pneumonia and septicaemia in humans. These diseases are frequently associated with thromboembolic cardiovascular complications. Pneumococci induce the exocytosis of endothelial Weibel-Palade Bodies and thereby actively stimulate the release of von Willebrand factor (VWF), which is an essential glycoprotein of the vascular hemostasis. Both, the pneumococcus induced pulmonary inflammation and the thromboembolytic complications are characterized by a dysbalanced hemostasis including a marked increase in VWF plasma concentrations. Here, we describe for the first time VWF as a novel interaction partner of capsulated and non-encapsulated pneumococci. Moreover, cell culture infection analyses with primary endothelial cells characterized VWF as bridging molecule that mediates bacterial adherence to endothelial cells in a heparin-sensitive manner. Due to the mechanoresponsive changes of the VWF protein conformation and multimerization status, which occur in the blood stream, we used a microfluidic pump system to generate shear flow-induced multimeric VWF strings on endothelial cell surfaces and analyzed attachment of RFP-expressing pneumococci in flow. By applying immunofluorescence visualization and additional electron microscopy, we detected a frequent and enduring bacterial attachment to the VWF strings. Bacterial attachment to the endothelium was confirmed in vivo using a zebrafish infection model, which is described in many reports and acknowledged as suitable model to study hemostasis mechanisms and protein interactions of coagulation factors. Notably, we visualized the recruitment of zebrafish-derived VWF to the surface of pneumococci circulating in the blood stream and detected a VWF-dependent formation of bacterial aggregates within the vasculature of infected zebrafish larvae. Furthermore, we identified the surface-exposed bacterial enolase as pneumococcal VWF binding protein, which interacts with the VWF domain A1 and determined the binding kinetics by surface plasmon resonance. Subsequent epitope mapping using an enolase peptide array indicates that the peptide 181YGAEIFHALKKILKS195 might serve as a possible core sequence of the VWF interaction site. In conclusion, we describe a VWF-mediated mechanism for pneumococcal anchoring within the bloodstream via surface-displayed enolase, which promotes intravascular bacterial aggregation.
Collapse
Affiliation(s)
- Hilger Jagau
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Ina-Kristin Behrens
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Karen Lahme
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Georgina Lorz
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Reinhard W Köster
- Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Reinhard Schneppenheim
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf (UKE Hamburg), Hamburg, Germany
| | - Tobias Obser
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf (UKE Hamburg), Hamburg, Germany
| | - Maria A Brehm
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf (UKE Hamburg), Hamburg, Germany
| | - Gesa König
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf (UKE Hamburg), Hamburg, Germany
| | - Thomas P Kohler
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Universität Greifswald, Greifswald, Germany
| | - Manfred Rohde
- Helmholtz Centre for Infection Research, Central Facility for Microscopy, Braunschweig, Germany
| | - Ronald Frank
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Werner Tegge
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Marcus Fulde
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Universität Greifswald, Greifswald, Germany
| | - Michael Steinert
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Molecular Infection Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Simone Bergmann
- Institute of Microbiology, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
14
|
Rosowski EE, Knox BP, Archambault LS, Huttenlocher A, Keller NP, Wheeler RT, Davis JM. The Zebrafish as a Model Host for Invasive Fungal Infections. J Fungi (Basel) 2018; 4:jof4040136. [PMID: 30551557 PMCID: PMC6308935 DOI: 10.3390/jof4040136] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 12/14/2022] Open
Abstract
The zebrafish has become a widely accepted model host for studies of infectious disease, including fungal infections. The species is genetically tractable, and the larvae are transparent and amenable to prolonged in vivo imaging and small molecule screening. The aim of this review is to provide a thorough introduction into the published studies of fungal infection in the zebrafish and the specific ways in which this model has benefited the field. In doing so, we hope to provide potential new zebrafish researchers with a snapshot of the current toolbox and prior results, while illustrating how the model has been used well and where the unfulfilled potential of this model can be found.
Collapse
Affiliation(s)
- Emily E Rosowski
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53716, USA.
| | - Benjamin P Knox
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53716, USA.
| | - Linda S Archambault
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA.
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53716, USA.
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53792, USA.
| | - Nancy P Keller
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53716, USA.
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Robert T Wheeler
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA.
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA.
| | - J Muse Davis
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
15
|
Csl2, a novel chimeric bacteriophage lysin to fight infections caused by Streptococcus suis, an emerging zoonotic pathogen. Sci Rep 2017; 7:16506. [PMID: 29184097 PMCID: PMC5705598 DOI: 10.1038/s41598-017-16736-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/16/2017] [Indexed: 12/20/2022] Open
Abstract
Streptococcus suis is a Gram-positive bacterium that infects humans and various animals, causing human mortality rates ranging from 5 to 20%, as well as important losses for the swine industry. In addition, there is no effective vaccine for S. suis and isolates with increasing antibiotic multiresistance are emerging worldwide. Facing this situation, wild type or engineered bacteriophage lysins constitute a promising alternative to conventional antibiotics. In this study, we have constructed a new chimeric lysin, Csl2, by fusing the catalytic domain of Cpl-7 lysozyme to the CW_7 repeats of LySMP lysin from an S. suis phage. Csl2 efficiently kills different S. suis strains and shows noticeable activity against a few streptococci of the mitis group. Specifically, 15 µg/ml Csl2 killed 4.3 logs of S. suis serotype 2 S735 strain in 60 min, in a buffer containing 150 mM NaCl and 10 mM CaCl2, at pH 6.0. We have set up a protocol to form a good biofilm with the non-encapsulated S. suis mutant strain BD101, and the use of 30 µg/ml Csl2 was enough for dispersing such biofilms and reducing 1–2 logs the number of planktonic bacteria. In vitro results have been validated in an adult zebrafish model of infection.
Collapse
|
16
|
Euba B, López-López N, Rodríguez-Arce I, Fernández-Calvet A, Barberán M, Caturla N, Martí S, Díez-Martínez R, Garmendia J. Resveratrol therapeutics combines both antimicrobial and immunomodulatory properties against respiratory infection by nontypeable Haemophilus influenzae. Sci Rep 2017; 7:12860. [PMID: 29038519 PMCID: PMC5643544 DOI: 10.1038/s41598-017-13034-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/12/2017] [Indexed: 12/21/2022] Open
Abstract
The respiratory pathogen nontypeable Haemophilus influenzae (NTHi) is an important cause of acute exacerbation of chronic obstructive pulmonary disease (AECOPD) that requires efficient treatments. A previous screening for host genes differentially expressed upon NTHi infection identified sirtuin-1, which encodes a NAD-dependent deacetylase protective against emphysema and is activated by resveratrol. This polyphenol concomitantly reduces NTHi viability, therefore highlighting its therapeutic potential against NTHi infection at the COPD airway. In this study, resveratrol antimicrobial effect on NTHi was shown to be bacteriostatic and did not induce resistance development in vitro. Analysis of modulatory properties on the NTHi-host airway epithelial interplay showed that resveratrol modulates bacterial invasion but not subcellular location, reduces inflammation without targeting phosphodiesterase 4B gene expression, and dampens β defensin-2 gene expression in infected cells. Moreover, resveratrol therapeutics against NTHi was evaluated in vivo on mouse respiratory and zebrafish septicemia infection model systems, showing to decrease NTHi viability in a dose-dependent manner and reduce airway inflammation upon infection, and to have a significant bacterial clearing effect without signs of host toxicity, respectively. This study presents resveratrol as a therapeutic of particular translational significance due to the attractiveness of targeting both infection and overactive inflammation at the COPD airway.
Collapse
Affiliation(s)
- Begoña Euba
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Instituto de Agrobiotecnología, CSIC-Universidad Pública Navarra-Gobierno Navarra, Mutilva, Spain
| | - Nahikari López-López
- Instituto de Agrobiotecnología, CSIC-Universidad Pública Navarra-Gobierno Navarra, Mutilva, Spain
| | - Irene Rodríguez-Arce
- Instituto de Agrobiotecnología, CSIC-Universidad Pública Navarra-Gobierno Navarra, Mutilva, Spain
| | - Ariadna Fernández-Calvet
- Instituto de Agrobiotecnología, CSIC-Universidad Pública Navarra-Gobierno Navarra, Mutilva, Spain
| | | | - Nuria Caturla
- Monteloeder, Elche Parque Empresarial, Elche, Alicante, Spain
| | - Sara Martí
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Departamento Microbiología, Hospital Universitari Bellvitge, University of Barcelona, IDIBELL, Barcelona, Spain
| | - Roberto Díez-Martínez
- Ikan Biotech SL, The Zebrafish Lab, Centro Europeo de Empresas e Innovación de Navarra (CEIN), Noáin, Spain
| | - Junkal Garmendia
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain. .,Instituto de Agrobiotecnología, CSIC-Universidad Pública Navarra-Gobierno Navarra, Mutilva, Spain.
| |
Collapse
|
17
|
Comprehensive validation of T- and B-cell deficiency in rag1-null zebrafish: Implication for the robust innate defense mechanisms of teleosts. Sci Rep 2017; 7:7536. [PMID: 28790360 PMCID: PMC5548773 DOI: 10.1038/s41598-017-08000-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 07/06/2017] [Indexed: 11/08/2022] Open
Abstract
rag1−/− zebrafish have been employed in immunological research as a useful immunodeficient vertebrate model, but with only fragmentary evidence for the lack of functional adaptive immunity. rag1-null zebrafish exhibit differences from their human and murine counterparts in that they can be maintained without any specific pathogen-free conditions. To define the immunodeficient status of rag1−/− zebrafish, we obtained further functional evidence on T- and B-cell deficiency in the fish at the protein, cellular, and organism levels. Our developed microscale assays provided evidence that rag1−/− fish do not possess serum IgM protein, that they do not achieve specific protection even after vaccination, and that they cannot induce antigen-specific CTL activity. The mortality rate in non-vaccinated fish suggests that rag1−/− fish possess innate protection equivalent to that of rag1+/− fish. Furthermore, poly(I:C)-induced immune responses revealed that the organ that controls anti-viral immunity is shifted from the spleen to the hepatopancreas due to the absence of T- and B-cell function, implying that immune homeostasis may change to an underside mode in rag-null fish. These findings suggest that the teleost relies heavily on innate immunity. Thus, this model could better highlight innate immunity in animals that lack adaptive immunity than mouse models.
Collapse
|
18
|
From Immunologically Archaic to Neoteric Glycovaccines. Vaccines (Basel) 2017; 5:vaccines5010004. [PMID: 28134792 PMCID: PMC5371740 DOI: 10.3390/vaccines5010004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/14/2016] [Accepted: 01/22/2017] [Indexed: 12/13/2022] Open
Abstract
Polysaccharides (PS) are present in the outermost surface of bacteria and readily come in contact with immune cells. They interact with specific antibodies, which in turn confer protection from infections. Vaccines with PS from pneumococci, meningococci, Haemophilus influenzae type b, and Salmonella typhi may be protective, although with the important constraint of failing to generate permanent immunological memory. This limitation has in part been circumvented by conjugating glycovaccines to proteins that stimulate T helper cells and facilitate the establishment of immunological memory. Currently, protection evoked by conjugated PS vaccines lasts for a few years. The same approach failed with PS from staphylococci, Streptococcus agalactiae, and Klebsiella. All those germs cause severe infections in humans and often develop resistance to antibiotic therapy. Thereby, prevention is of increasing importance to better control outbreaks. As only 23 of more than 90 pneumococcal serotypes and 4 of 13 clinically relevant Neisseria meningitidis serogroups are covered by available vaccines there is still tremendous clinical need for PS vaccines. This review focuses on glycovaccines and the immunological mechanisms for their success or failure. We discuss recent advances that may facilitate generation of high affinity anti-PS antibodies and confer specific immunity and long-lasting protection.
Collapse
|
19
|
Sullivan C, Matty MA, Jurczyszak D, Gabor KA, Millard PJ, Tobin DM, Kim CH. Infectious disease models in zebrafish. Methods Cell Biol 2016; 138:101-136. [PMID: 28129840 DOI: 10.1016/bs.mcb.2016.10.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In recent years, the zebrafish (Danio rerio) has developed as an important alternative to mammalian models for the study of hostpathogen interactions. Because they lack a functional adaptive immune response during the first 4-6weeks of development, zebrafish rely upon innate immune responses to protect against injuries and infections. During this early period of development, it is possible to isolate and study mechanisms of infection and inflammation arising from the innate immune response without the complications presented by the adaptive immune response. Zebrafish possess several inherent characteristics that make them an attractive option to study hostpathogen interactions, including extensive sequence and functional conservation with the human genome, optical clarity in larvae that facilitates the high-resolution visualization of host cell-microbe interactions, a fully sequenced and annotated genome, robust forward and reverse genetic tools and techniques (e.g., CRISPR-Cas9 and TALENs), and amenability to chemical studies and screens. Here, we describe methods for studying hostpathogen interactions both through systemic infections and through localized infections that allow analysis of host cell response, migration patterns, and behavior. Each of the methods described can be modified for use in downstream applications that include ecotoxicant studies and chemical screens.
Collapse
Affiliation(s)
- C Sullivan
- University of Maine, Orono, ME, United States
| | - M A Matty
- Duke University School of Medicine, Durham, NC, United States
| | | | - K A Gabor
- National Institute of Environmental Health Sciences, Durham, NC, United States
| | - P J Millard
- University of Maine, Orono, ME, United States
| | - D M Tobin
- Duke University School of Medicine, Durham, NC, United States
| | - C H Kim
- University of Maine, Orono, ME, United States
| |
Collapse
|
20
|
Jim KK, Engelen-Lee J, van der Sar AM, Bitter W, Brouwer MC, van der Ende A, Veening JW, van de Beek D, Vandenbroucke-Grauls CMJE. Infection of zebrafish embryos with live fluorescent Streptococcus pneumoniae as a real-time pneumococcal meningitis model. J Neuroinflammation 2016; 13:188. [PMID: 27542968 PMCID: PMC4992281 DOI: 10.1186/s12974-016-0655-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/08/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Streptococcus pneumoniae is one of the most important causes of bacterial meningitis, an infection where unfavourable outcome is driven by bacterial and host-derived toxins. In this study, we developed and characterized a pneumococcal meningitis model in zebrafish embryos that allows for real-time investigation of early host-microbe interaction. METHODS Zebrafish embryos were infected in the caudal vein or hindbrain ventricle with green fluorescent wild-type S. pneumoniae D39 or a pneumolysin-deficient mutant. The kdrl:mCherry transgenic zebrafish line was used to visualize the blood vessels, whereas phagocytic cells were visualized by staining with far red anti-L-plastin or in mpx:GFP/mpeg1:mCherry zebrafish, that have green fluorescent neutrophils and red fluorescent macrophages. Imaging was performed by fluorescence confocal and time-lapse microscopy. RESULTS After infection by caudal vein, we saw focal clogging of the pneumococci in the blood vessels and migration of bacteria through the blood-brain barrier into the subarachnoid space and brain tissue. Infection with pneumolysin-deficient S. pneumoniae in the hindbrain ventricle showed attenuated growth and migration through the brain as compared to the wild-type strain. Time-lapse and confocal imaging revealed that the initial innate immune response to S. pneumoniae in the subarachnoid space mainly consisted of neutrophils and that pneumolysin-mediated cytolytic activity caused a marked reduction of phagocytes. CONCLUSIONS This new meningitis model permits detailed analysis and visualization of host-microbe interaction in pneumococcal meningitis in real time and is a very promising tool to further our insights in the pathogenesis of pneumococcal meningitis.
Collapse
Affiliation(s)
- Kin Ki Jim
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Department of Neurology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - JooYeon Engelen-Lee
- Department of Neurology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Astrid M van der Sar
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Wilbert Bitter
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Matthijs C Brouwer
- Department of Neurology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Arie van der Ende
- Department of Medical Microbiology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- The Netherlands Reference Laboratory for Bacterial Meningitis, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jan-Willem Veening
- Molecular Genetics Group, Groningen Biomolecular Sciences and Biotechnology Institute, Centre for Synthetic Biology, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Diederik van de Beek
- Department of Neurology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Christina M J E Vandenbroucke-Grauls
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands.
- Department of Medical Microbiology and Infection Control, VU University Medical Center, P.O. Box 7057, 1007 MB, Amsterdam, The Netherlands.
| |
Collapse
|
21
|
Saralahti A, Rämet M. Zebrafish and Streptococcal Infections. Scand J Immunol 2015; 82:174-83. [PMID: 26095827 DOI: 10.1111/sji.12320] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 04/28/2015] [Indexed: 12/11/2022]
Abstract
Streptococcal bacteria are a versatile group of gram-positive bacteria capable of infecting several host organisms, including humans and fish. Streptococcal species are common colonizers of the human respiratory and gastrointestinal tract, but they also cause some of the most common life-threatening, invasive infections in humans and aquaculture. With its unique characteristics and efficient tools for genetic and imaging applications, the zebrafish (Danio rerio) has emerged as a powerful vertebrate model for infectious diseases. Several zebrafish models introduced so far have shown that zebrafish are suitable models for both zoonotic and human-specific infections. Recently, several zebrafish models mimicking human streptococcal infections have also been developed. These models show great potential in providing novel information about the pathogenic mechanisms and host responses associated with human streptococcal infections. Here, we review the zebrafish infection models for the most relevant streptococcal species: the human-specific Streptococcus pneumoniae and Streptococcus pyogenes, and the zoonotic Streptococcus iniae and Streptococcus agalactiae. The recent success and the future potential of these models for the study of host-pathogen interactions in streptococcal infections are also discussed.
Collapse
Affiliation(s)
- A Saralahti
- BioMediTech, University of Tampere, Tampere, Finland
| | - M Rämet
- BioMediTech, University of Tampere, Tampere, Finland.,Department of Pediatrics, Tampere University Hospital, Tampere, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland.,PEDEGO Research Center, Medical Research Center Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
22
|
Harvie EA, Huttenlocher A. Neutrophils in host defense: new insights from zebrafish. J Leukoc Biol 2015; 98:523-37. [PMID: 25717145 PMCID: PMC4569048 DOI: 10.1189/jlb.4mr1114-524r] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 01/15/2015] [Accepted: 01/19/2015] [Indexed: 12/11/2022] Open
Abstract
Neutrophils are highly motile phagocytic cells that play a critical role in the immune response to infection. Zebrafish (Danio rerio) are increasingly used to study neutrophil function and host-pathogen interactions. The generation of transgenic zebrafish lines with fluorescently labeled leukocytes has made it possible to visualize the neutrophil response to infection in real time by use of optically transparent zebrafish larvae. In addition, the genetic tractability of zebrafish has allowed for the generation of models of inherited neutrophil disorders. In this review, we discuss several zebrafish models of infectious disease, both in the context of immunocompetent, as well as neutrophil-deficient hosts and how these models have shed light on neutrophil behavior during infection.
Collapse
Affiliation(s)
- Elizabeth A Harvie
- *Microbiology Doctoral Training Program, Departments of Medical Microbiology and Immunology and Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Anna Huttenlocher
- *Microbiology Doctoral Training Program, Departments of Medical Microbiology and Immunology and Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
23
|
López Hernández Y, Yero D, Pinos-Rodríguez JM, Gibert I. Animals devoid of pulmonary system as infection models in the study of lung bacterial pathogens. Front Microbiol 2015; 6:38. [PMID: 25699030 PMCID: PMC4316775 DOI: 10.3389/fmicb.2015.00038] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 01/12/2015] [Indexed: 01/15/2023] Open
Abstract
Biological disease models can be difficult and costly to develop and use on a routine basis. Particularly, in vivo lung infection models performed to study lung pathologies use to be laborious, demand a great time and commonly are associated with ethical issues. When infections in experimental animals are used, they need to be refined, defined, and validated for their intended purpose. Therefore, alternative and easy to handle models of experimental infections are still needed to test the virulence of bacterial lung pathogens. Because non-mammalian models have less ethical and cost constraints as a subjects for experimentation, in some cases would be appropriated to include these models as valuable tools to explore host-pathogen interactions. Numerous scientific data have been argued to the more extensive use of several kinds of alternative models, such as, the vertebrate zebrafish (Danio rerio), and non-vertebrate insects and nematodes (e.g., Caenorhabditis elegans) in the study of diverse infectious agents that affect humans. Here, we review the use of these vertebrate and non-vertebrate models in the study of bacterial agents, which are considered the principal causes of lung injury. Curiously none of these animals have a respiratory system as in air-breathing vertebrates, where respiration takes place in lungs. Despite this fact, with the present review we sought to provide elements in favor of the use of these alternative animal models of infection to reveal the molecular signatures of host-pathogen interactions.
Collapse
Affiliation(s)
- Yamilé López Hernández
- Centro de Biociencias, Universidad Autónoma de San Luis Potosí San Luis de Potosí, Mexico
| | - Daniel Yero
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona Barcelona, Spain ; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona Barcelona, Spain
| | - Juan M Pinos-Rodríguez
- Centro de Biociencias, Universidad Autónoma de San Luis Potosí San Luis de Potosí, Mexico
| | - Isidre Gibert
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona Barcelona, Spain ; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona Barcelona, Spain
| |
Collapse
|