1
|
Papadopoulos S, Hardy D, Vernel-Pauillac F, Tichit M, Boneca IG, Werts C. Myocarditis and neutrophil-mediated vascular leakage but not cytokine storm associated with fatal murine leptospirosis. EBioMedicine 2025; 112:105571. [PMID: 39889371 PMCID: PMC11830356 DOI: 10.1016/j.ebiom.2025.105571] [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: 08/18/2024] [Revised: 01/03/2025] [Accepted: 01/15/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Leptospirosis is a globally neglected re-emerging zoonosis affecting all mammals, albeit with variable outcomes. Humans are susceptible to leptospirosis; infection with Leptospira interrogans species can cause severe disease in humans, with multi-organ failure, mainly affecting kidney, lung and liver function, leading to death in 10% of cases. Mice and rats are more resistant to acute disease and can carry leptospires asymptomatically in the kidneys and act as reservoirs, shedding leptospires into the environment. The incidence of leptospirosis is higher in tropical countries, and countries with poor sanitation, where heavy rainfall and flooding favour infection. Diagnosis of leptospirosis is difficult because of the many different serovars and the variety of clinical symptoms that can be confused with viral infections. The physiopathology is poorly understood, and leptospirosis is often regarded as an inflammatory disease, like sepsis. METHODS To investigate the causes of death in lethal leptospirosis, we compared intraperitoneal infection of male and female C57BL6/J mice with 108Leptospira of two strains of pathogenic L. interrogans. One strain, L. interrogans Manilae L495, killed the mice 4 days after infection, whereas the other strain, L. interrogans Icterohaemorrhagiae Verdun, did not induce any major symptoms in the mice. On day 3 post infection, the mice were humanely euthanised and blood and organs were collected. Bacterial load, biochemical parameters, cytokine production and leucocyte population were assessed by qPCR, ELISA, cytometry and immunohistochemistry. FINDINGS Neither lung, liver, pancreas or kidney damage nor massive necroptosis or cytokine storm could explain the lethality. Although we did not find pro-inflammatory cytokines, we did find elevated levels of the anti-inflammatory cytokine IL-10 and the chemokine RANTES in the serum and organs of Leptospira-infected mice. In contrast, severe leptospirosis was associated with neutrophilia and vascular permeability, unexpectedly due to neutrophils and not only due to Leptospira infection. Strikingly, the main cause of death was myocarditis, an overlooked complication of human leptospirosis. INTERPRETATION Despite clinical similarities between bacterial sepsis and leptospirosis, striking differences were observed, in particular a lack of cytokine storm in acute leptospirosis. The fact that IL-10 was increased in infected mice may explain the lack of pro-inflammatory cytokines, emphasising the covert nature of Leptospira infections. Neutrophilia is a hallmark of human leptospirosis. Our findings confirm the ineffective control of infection by neutrophils and highlight their deleterious role in vascular permeability, previously only attributed to the ability of leptospires to damage and cross endothelial junctions. Finally, the identification of death due to myocarditis rather than kidney, liver or liver failure may reflect an overlooked but common symptom associated with poor prognosis in human leptospirosis. These features of neutrophilia and myocarditis are also seen in patients, making this mouse model a paradigm for better understanding human leptospirosis and designing new therapeutic strategies. FUNDING The Boneca laboratory was supported by the following programmes: Investissement d'Avenir program, Laboratoire d'Excellence "Integrative Biology of Emerging Infectious Diseases" (ANR-10-LABX-62-IBEID) and by R&D grants from Danone and MEIJI. CW received an ICRAD/ANR grant (S-CR23012-ANR 22 ICRD 0004 01). SP received a scholarship by Université Paris Cité (formerly Université Paris V - Descartes) through Doctoral School BioSPC (ED562, BioSPC). SP has additionally received a scholarship "Fin de Thèse de Science" number FDT202404018322 granted by "Fondation pour la Recherche Médicale (FRM)". The funders had no implication in the design, analysis and reporting of the study.
Collapse
Affiliation(s)
- Stylianos Papadopoulos
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, F-75015, France
| | - David Hardy
- Institut Pasteur, Université Paris Cité, Histopathology Core Facility, Paris, F-75015, France
| | - Frédérique Vernel-Pauillac
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, F-75015, France
| | - Magali Tichit
- Institut Pasteur, Université Paris Cité, Histopathology Core Facility, Paris, F-75015, France
| | - Ivo G Boneca
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, F-75015, France
| | - Catherine Werts
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, F-75015, France.
| |
Collapse
|
2
|
Liu C, Shao J, Ma X, Tang Y, Li J, Li H, Chi X, Liu Z. A novel two-component system contributing the catabolism of c-di-GMP influences virulence in Aeromonas veronii. Front Microbiol 2025; 16:1527317. [PMID: 39980697 PMCID: PMC11841396 DOI: 10.3389/fmicb.2025.1527317] [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: 11/13/2024] [Accepted: 01/16/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Response regulators from diverse two-component systems often function as diguanylate cyclases or phosphodiesterases, thereby enabling precise regulation of intracellular c-di-GMP levels to control bacterial virulence and motility. However, the regulatory mechanisms of c-di-GMP require further elucidation. Methods This study confirmed that ArrS and ArrR form a two-component system via structural analysis, two-hybrid, and phosphodiesterase activity detection. To evaluate the impact of ArrS/ArrR on intracellular c-di-GMP levels, biofilm detection, motility detection, fluorescence reporter plasmids, and LC-MS/MS analysis were employed. One-hybrid, EMSA, and RT-qPCR were used to demonstrate the function of ArgR on arrSR promoter. The roles of ArrS/ArrR in Aeromonas veronii were investigated using RT-qPCR, murine model, and proteomics. Results ArrS and ArrR constituted a two-component system in Aeromonas veronii and were transcriptionally repressed by ArgR. ArrR exhibited phosphodiesterase activity, which is inhibited through phosphorylation mediated by ArrS. In Aeromonas veronii, ArrS/ArrR significantly altered the intracellular c-di-GMP levels. In a murine model, ΔarrS exhibited increased pathogenicity, leading to elevated TNF-α and IFN-γ levels in serum, and severer toxicity to spleen and kidney. These effects might be elucidated by the upregulated inflammation-associated proteins in ΔarrS. Moreover, the exonuclease RecB was also up-regulated in ΔarrS. Discussion We elucidated the regulatory mechanism of ArrS/ArrR on intracellular c-di-GMP levels and its impact on the virulence in Aeromonas veronii, and discussed the intricate relationship between c-di-GMP metabolism and arginine metabolism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhu Liu
- School of Life and Health Sciences, Hainan University, Haikou, China
| |
Collapse
|
3
|
Moustafa MAM, Schlachter S, Parveen N. Innovative Strategies to Study the Pathogenesis of Elusive Spirochetes and Difficulties Managing the Chronic Infections They Cause. Annu Rev Microbiol 2024; 78:337-360. [PMID: 39107040 DOI: 10.1146/annurev-micro-100423-030847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2024]
Abstract
The major human spirochetal pathogens (Leptospira, Borrelia, and Treponema) are difficult to diagnose and lack vaccines to prevent infections. Infection by these spirochetes does not generate general protective immunity, allowing reinfection by different strains to occur. These stealth pathogens have uncommon physiology, pathogenesis, and clinical presentations and possess unique immune evasion mechanisms to facilitate their host adaptation and persistence. Collectively, host-spirochete interactions orchestrate systemic infections in a manner distinct from organ- and tissue-specific diseases caused by many bacterial pathogens. Difficulties in growing and genetic manipulation of infectious spirochetes have hindered the full understanding of their virulence factors despite decades to centuries of research. This article highlights the current understanding of the intricacies of spirochetal pathogenesis and diseases. Our comprehensive review of the progress versus gaps in knowledge lays a foundation for researchers to direct their studies toward the development of effective diagnostics and vaccines to protect patients from serious, chronic spirochetal diseases.
Collapse
Affiliation(s)
| | - Samantha Schlachter
- Department of Biology, Saint Elizabeth University, Morristown, New Jersey, USA
| | - Nikhat Parveen
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey, USA;
| |
Collapse
|
4
|
Xie X, Chen X, Zhang S, Liu J, Zhang W, Cao Y. Neutralizing gut-derived lipopolysaccharide as a novel therapeutic strategy for severe leptospirosis. eLife 2024; 13:RP96065. [PMID: 38818711 PMCID: PMC11142641 DOI: 10.7554/elife.96065] [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: 06/01/2024] Open
Abstract
Leptospirosis is an emerging infectious disease caused by pathogenic Leptospira spp. Humans and some mammals can develop severe forms of leptospirosis accompanied by a dysregulated inflammatory response, which often results in death. The gut microbiota has been increasingly recognized as a vital element in systemic health. However, the precise role of the gut microbiota in severe leptospirosis is still unknown. Here, we aimed to explore the function and potential mechanisms of the gut microbiota in a hamster model of severe leptospirosis. Our study showed that leptospires were able to multiply in the intestine, cause pathological injury, and induce intestinal and systemic inflammatory responses. 16S rRNA gene sequencing analysis revealed that Leptospira infection changed the composition of the gut microbiota of hamsters with an expansion of Proteobacteria. In addition, gut barrier permeability was increased after infection, as reflected by a decrease in the expression of tight junctions. Translocated Proteobacteria were found in the intestinal epithelium of moribund hamsters, as determined by fluorescence in situ hybridization, with elevated lipopolysaccharide (LPS) levels in the serum. Moreover, gut microbiota depletion reduced the survival time, increased the leptospiral load, and promoted the expression of proinflammatory cytokines after Leptospira infection. Intriguingly, fecal filtration and serum from moribund hamsters both increased the transcription of TNF-α, IL-1β, IL-10, and TLR4 in macrophages compared with those from uninfected hamsters. These stimulating activities were inhibited by LPS neutralization using polymyxin B. Based on our findings, we identified an LPS neutralization therapy that significantly improved the survival rates in severe leptospirosis when used in combination with antibiotic therapy or polyclonal antibody therapy. In conclusion, our study not only uncovers the role of the gut microbiota in severe leptospirosis but also provides a therapeutic strategy for severe leptospirosis.
Collapse
Affiliation(s)
- Xufeng Xie
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin UniversityJilinChina
| | - Xi Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin UniversityJilinChina
| | - Shilei Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin UniversityJilinChina
| | - Jiuxi Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin UniversityJilinChina
| | - Wenlong Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin UniversityJilinChina
| | - Yongguo Cao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin UniversityJilinChina
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin UniversityChangchunChina
| |
Collapse
|
5
|
Chisholm LO, Jaeger NM, Murawsky HE, Harms MJ. S100A9 interacts with a dynamic region on CD14 to activate Toll-like receptor 4. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594416. [PMID: 38798518 PMCID: PMC11118535 DOI: 10.1101/2024.05.15.594416] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
S100A9 is a Damage Associated Molecular Pattern (DAMP) that activates inflammatory pathways via Toll-like receptor 4 (TLR4). This activity plays important homeostatic roles in tissue repair, but can also contribute to inflammatory diseases. The mechanism of activation is unknown. Here, we follow up on a previous observation that the protein CD14 is an important co-receptor that enables S100A9 to activate TLR4. Using cell-based functional assays and a combination of mutations and pharmocological perturbations, we found that CD14 must be membrane bound to potentiate TLR4 activation by S100A9. Additionally, S100A9 is sensitive to inhibitors of pathways downstream of TLR4 internalization. Together, this suggests that S100A9 induces activity via CD14-dependent internalization of TLR4. We then used mutagenesis, structural modeling, and in vitro binding experiments to establish that S100A9 binds to CD14's N-terminus in a region that overlaps with, but is not identical to, the region where CD14 binds its canonical ligand, lipopolysaccharide (LPS). In molecular dynamics simulations, this region of the protein is dynamic, allowing it to reorganize to recognize both S100A9 (a soluble protein) and LPS (a small hydrophobic molecule). Our work is the first attempt at a molecular characterization of the S100A9/CD14 interaction, bringing us one step closer to unraveling the full mechanism by which S100A9 activates TLR4/MD-2.
Collapse
|
6
|
Bonhomme D, Santecchia I, Escoll P, Papadopoulos S, Vernel-Pauillac F, Boneca IG, Werts C. Leptospiral lipopolysaccharide dampens inflammation through upregulation of autophagy adaptor p62 and NRF2 signaling in macrophages. Microbes Infect 2024; 26:105274. [PMID: 38081475 DOI: 10.1016/j.micinf.2023.105274] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Leptospira interrogans are pathogenic bacteria responsible for leptospirosis, a worldwide zoonosis. All vertebrates can be infected, and some species like humans are susceptible to the disease whereas rodents such as mice are resistant and become asymptomatic renal carriers. Leptospires are stealth bacteria that are known to escape several immune recognition pathways and resist killing mechanisms. We recently published that leptospires may survive intracellularly in and exit macrophages, avoiding xenophagy, a pathogen-targeting form of autophagy. Interestingly, the latter is one of the antimicrobial mechanisms often highjacked by bacteria to evade the host immune response. In this study we explored whether leptospires subvert the key molecular players of autophagy to facilitate infection. We showed in macrophages that leptospires triggered a specific accumulation of autophagy-adaptor p62 in puncta-like structures, without altering autophagic flux. We demonstrated that Leptospira-induced p62 accumulation is a passive mechanism depending on the leptospiral virulence factor LPS signaling via TLR4/TLR2. p62 is a central pleiotropic protein, also mediating cell stress and death, via the translocation of transcription factors. We demonstrated that Leptospira-driven accumulation of p62 induced the translocation of transcription factor NRF2, a key player in the anti-oxidant response. However, NRF2 translocation upon Leptospira infection did not result as expected in antioxydant response, but dampened the production of inflammatory mediators such as iNOS/NO, TNF and IL6. Overall, these findings highlight a novel passive bacterial mechanism linked to LPS and p62/NRF2 signaling that decreases inflammation and contributes to the stealthiness of leptospires.
Collapse
Affiliation(s)
- Delphine Bonhomme
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Ignacio Santecchia
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Pedro Escoll
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité Biologie des Bactéries Intracellulaires, Paris, France
| | - Stylianos Papadopoulos
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Frédérique Vernel-Pauillac
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Ivo G Boneca
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Catherine Werts
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France.
| |
Collapse
|
7
|
Bonhomme D, Cavaillon JM, Werts C. The dangerous liaisons in innate immunity involving recombinant proteins and endotoxins: Examples from the literature and the Leptospira field. J Biol Chem 2024; 300:105506. [PMID: 38029965 PMCID: PMC10777017 DOI: 10.1016/j.jbc.2023.105506] [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: 07/24/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/01/2023] Open
Abstract
Endotoxins, also known as lipopolysaccharides (LPS), are essential components of cell walls of diderm bacteria such as Escherichia coli. LPS are microbe-associated molecular patterns that can activate pattern recognition receptors. While trying to investigate the interactions between proteins and host innate immunity, some studies using recombinant proteins expressed in E. coli reported interaction and activation of immune cells. Here, we set out to provide information on endotoxins that are highly toxic to humans and bind to numerous molecules, including recombinant proteins. We begin by outlining the history of the discovery of endotoxins, their receptors and the associated signaling pathways that confer extreme sensitivity to immune cells, acting alone or in synergy with other microbe-associated molecular patterns. We list the various places where endotoxins have been found. Additionally, we warn against the risk of data misinterpretation due to endotoxin contamination in recombinant proteins, which is difficult to estimate with the Limulus amebocyte lysate assay, and cannot be completely neutralized (e.g., treatment with polymyxin B or heating). We further illustrate our point with examples of recombinant heat-shock proteins and viral proteins from severe acute respiratory syndrome coronavirus 2, dengue and HIV, for which endotoxin contamination has eventually been shown to be responsible for the inflammatory roles previously ascribed. We also critically appraised studies on recombinant Leptospira proteins regarding their putative inflammatory roles. Finally, to avoid these issues, we propose alternatives to express recombinant proteins in nonmicrobial systems. Microbiologists wishing to undertake innate immunity studies with their favorite pathogens should be aware of these difficulties.
Collapse
Affiliation(s)
- Delphine Bonhomme
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | | | - Catherine Werts
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France.
| |
Collapse
|
8
|
Varma VP, Bankala R, Kumar A, Gawai S, Faisal SM. Differential modulation of innate immune response by lipopolysaccharide of Leptospira. Open Biol 2023; 13:230101. [PMID: 37935355 PMCID: PMC10645091 DOI: 10.1098/rsob.230101] [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: 05/30/2023] [Accepted: 08/16/2023] [Indexed: 11/09/2023] Open
Abstract
Leptospirosis is a worldwide zoonosis caused by pathogenic Leptospira spp. having more than 300 serovars. These serovars can infect a variety of hosts, some being asymptomatic carriers and others showing varied symptoms of mild to severe infection. Since lipopolysaccharide (LPS) is the major antigen which defines serovar specificity, this different course of infection may be attributed to a differential innate response against this antigen. Previous studies have shown that Leptospira LPS is less endotoxic. However, it is unclear whether there is a difference in the ability of LPS isolated from different serovars to modulate the innate response. In this study, we purified LPS from three widely prevalent pathogenic serovars, i.e. Icterohaemorrhagiae strain RGA, Pomona, Hardjo, and from non-pathogenic L. biflexa serovar semeranga strain Potac 1 collectively termed as L-LPS and tested their ability to modulate innate response in macrophages from both resistant (mice) and susceptible (human and bovine) hosts. L-LPS induced differential response being more proinflammatory in mouse and less proinflammatory in human and bovine macrophages but overall less immunostimulatory than E. coli LPS (E-LPS). Irrespective of serovar, this response was TLR2-dependent in humans, whereas TLR4-dependent/CD14-independent in mouse using MyD88 adapter and signalling through P38 and ERK-dependent MAP kinase pathway. L-LPS-activated macrophages were able to phagocytose Leptospira and this effect was significantly higher or more pronounced when the macrophages were stimulated with L-LPS from the corresponding serovar. L-LPS activated both canonical and non-canonical inflammasome, producing IL-1β without inducing pyroptosis. Further, L-LPS induced both TNF-mediated early and NO-mediated late apoptosis. Altogether, these results indicate that L-LPS induces a differential innate response that is quite distinct from that induced by E-LPS and may be attributed to the structural differences and its atypical nature.
Collapse
Affiliation(s)
- Vivek P. Varma
- Laboratory of Vaccine Immunology, National Institute of Animal Biotechnology, Hyderabad 500032, India
- Graduate Studies, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Ramudu Bankala
- Laboratory of Vaccine Immunology, National Institute of Animal Biotechnology, Hyderabad 500032, India
| | - Ajay Kumar
- Laboratory of Vaccine Immunology, National Institute of Animal Biotechnology, Hyderabad 500032, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Shashikant Gawai
- Laboratory of Vaccine Immunology, National Institute of Animal Biotechnology, Hyderabad 500032, India
| | - Syed M. Faisal
- Laboratory of Vaccine Immunology, National Institute of Animal Biotechnology, Hyderabad 500032, India
- Regional Centre for Biotechnology, Faridabad, India
| |
Collapse
|
9
|
Azevedo IR, Amamura TA, Isaac L. Human leptospirosis: In search for a better vaccine. Scand J Immunol 2023; 98:e13316. [PMID: 39008520 DOI: 10.1111/sji.13316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/17/2024]
Abstract
Leptospirosis is a neglected disease caused by bacteria of the genus Leptospira and is more prevalent in tropical and subtropical countries. This pathogen infects humans and other animals, responsible for the most widespread zoonosis in the world, estimated to be responsible for 60 000 deaths and 1 million cases per year. To date, commercial vaccines against human leptospirosis are available only in some countries such as Japan, China, Cuba and France. These vaccines prepared with inactivated Leptospira (bacterins) induce a short-term and serovar-specific immune response, with strong adverse side effects. To circumvent these limitations, several research groups are investigating new experimental vaccines in order to ensure that they are safe, efficient, and protect against several pathogenic Leptospira serovars, inducing sterilizing immunity. Most of these protocols use attenuated cultures, preparations after LPS removal, recombinant proteins or DNA from pathogenic Leptospira spp. The aim of this review was to highlight several promising vaccine candidates, considering their immunogenicity, presence in different pathogenic Leptospira serovars, their role in virulence or immune evasion and other factors.
Collapse
Affiliation(s)
- Isabela Resende Azevedo
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Thais Akemi Amamura
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lourdes Isaac
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Gangani D, Dissanayake W, de Silva R, Anuradha K, Karunanayake L, Fernando N, Rajapakse S, Premawansa S, Handunnetti S. Humoral immune response and changes in peritoneal cell populations in rats immunized against two Leptospira serovars; serovar patoc and serovar pyrogenes. BMC Immunol 2023; 24:39. [PMID: 37848809 PMCID: PMC10583450 DOI: 10.1186/s12865-023-00574-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 10/09/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Leptospirosis is a zoonotic disease caused by Leptospira species. Variations in lipopolysaccharide (LPS) structure in Leptospira are known to be associated with the serovar diversity and antigenicity. Development of immunodiagnostics for early detection of leptospirosis based on immune responses against different pathogenic antigens as well as development of vaccines are important. Hence, this study has assessed the immune response generated against leptospiral LPS and whole antigen preparations of pathogenic and saprophytic Leptospira and specific changes in peritoneal cells was also studied to elucidate the cellular responses associated with immune response of Wistar rats. METHODS During the study, immune response induced by two types of Leptospira antigen preparations of two selected serovars was compared. Changes in the specific peritoneal cell subpopulations following immunizations of rats were analyzed using flow cytometry. RESULTS Of the two antigen preparations tested, the LPS extract induced a higher IgM immune response as opposed to the sonicated antigen preparation. Of the two serovars tested, L. interrogans serovar Pyrogenes had induced a higher IgM response compared to that by L. biflexa serovar Patoc. Considering the IgG titers, equivalent responses were observed with all four antigen preparations. Significant increases in lymphocytes were observed following immunization with LPS of both serovars. Interestingly, the B2 cell percentages increased significantly during the immunization period. Further, significant correlations were observed with both IgM and IgG responses and percentage of B2 cells in the peritoneal cavity (PC). CONCLUSION LPS extract of L. interrogans serovar Pyrogenes induced higher IgM response while the IgG response was equivalent among the four antigen preparations tested. Significant increase of B2 cell percentage in the peritoneal cavity during the immunization reflects the accumulation of B2 cells in the PC which may play considerable role in generating humoral response against Leptospira antigens.
Collapse
Affiliation(s)
- Dakshika Gangani
- Institute of Biochemistry Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Colombo, Sri Lanka.
| | - Wathsala Dissanayake
- Institute of Biochemistry Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Colombo, Sri Lanka
| | - Rajiva de Silva
- Department of Immunology, Medical Research Institute, Colombo 08, Colombo, Sri Lanka
| | - Kaushalya Anuradha
- Institute of Biochemistry Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Colombo, Sri Lanka
| | - Lilani Karunanayake
- Department of Bacteriology, Medical Research Institute, Colombo 08, Colombo, Sri Lanka
| | - Narmada Fernando
- Institute of Biochemistry Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Colombo, Sri Lanka
| | - Senaka Rajapakse
- Postgraduate Institute of Medicine, University of Colombo, Colombo 08, Colombo, Sri Lanka
| | - Sunil Premawansa
- Department of Zoology and Environment Sciences, Faculty of Science, University of Colombo, Colombo 03, Colombo, Sri Lanka
| | - Shiroma Handunnetti
- Institute of Biochemistry Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Colombo, Sri Lanka
| |
Collapse
|
11
|
Bonhomme D, Hernandez-Trejo V, Papadopoulos S, Pigache R, Fanton d'Andon M, Outlioua A, Boneca IG, Werts C. Leptospira interrogans Prevents Macrophage Cell Death and Pyroptotic IL-1β Release through Its Atypical Lipopolysaccharide. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:459-474. [PMID: 36602965 DOI: 10.4049/jimmunol.2200584] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/03/2022] [Indexed: 01/06/2023]
Abstract
Leptospira interrogans are bacteria that can infect all vertebrates and are responsible for leptospirosis, a neglected zoonosis. Some hosts, such as humans, are susceptible to the disease, whereas mice are resistant and get chronically colonized. Although leptospires escape recognition by some immune receptors, they activate the NOD-like receptor pyrin 3-inflammasome and trigger IL-1β secretion. Classically, IL-1β secretion is associated with lytic inflammatory cell death called pyroptosis, resulting from cytosolic LPS binding to inflammatory caspases, such as caspase 11. Interestingly, we showed that L. interrogans and Leptospira biflexa do not trigger cell death in either murine, human, hamster, or bovine macrophages, escaping both pyroptosis and apoptosis. We showed, in murine cells, that the mild IL-1β secretion induced by leptospires occurred through nonlytic caspase 8-dependent gasdermin D pore formation and not through activation of caspase 11/noncanonical inflammasome. Strikingly, we demonstrated a potent antagonistic effect of pathogenic L. interrogans and their atypical LPS on spontaneous and Escherichia coli LPS-induced cell death. Indeed, LPS of L. interrogans efficiently prevents caspase 11 dimerization and subsequent massive gasdermin D cleavage. Finally, we showed that pyroptosis escape by leptospires prevents massive IL-1β release, and we consistently found no major role of IL-1R in controlling experimental leptospirosis in vivo. Overall, to our knowledge, our findings described a novel mechanism by which leptospires dampen inflammation, thus potentially contributing to their stealthiness.
Collapse
Affiliation(s)
- Delphine Bonhomme
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Veronica Hernandez-Trejo
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Stylianos Papadopoulos
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Rémi Pigache
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Martine Fanton d'Andon
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Ahmed Outlioua
- INSERM, UMR_S 1197, Hôpital Paul Brousse, Villejuif, France.,Université Paris-Saclay, Paris, France; and.,Health and Environment Laboratory, Aïn Chock Faculty of Sciences, Hassan II University of Casablanca, Casablanca, Morocco
| | - Ivo G Boneca
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Catherine Werts
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| |
Collapse
|
12
|
Bai Y, Lyu M, Fukunaga M, Watanabe S, Iwatani S, Miyanaga K, Yamamoto N. Lactobacillus johnsonii enhances the gut barrier integrity via the interaction between GAPDH and the mouse tight junction protein JAM-2. Food Funct 2022; 13:11021-11033. [PMID: 36069670 DOI: 10.1039/d2fo00886f] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Commensal intestinal microbiota interacts with gut epithelial cells in the host by binding to specific host receptors. Several pattern recognition receptors on the gut that sense conserved microbial-associated molecular patterns have been reported; however, many of the gut receptor molecules involved in bacterial binding have not yet been identified. In this study, commensal intestinal bacteria interacting with mouse gut surface proteins were screened from fecal bacterial samples, to identify novel receptors on the epithelial cells in the mouse gut. Among the screened intestinal lactic acid bacteria, the frequently isolated Lactobacillus johnsonii MG was used for the purification of gut receptor proteins. An approximately 30 kDa protein was purified using affinity resin coupled surface layer proteins isolated from L. johnsonii MG. The purified gut protein was identified as a member of the tight junction protein family, junctional adhesion molecule-2 (JAM-2). As expected, the tight junctions of Caco-2 cells damaged by H2O2 were repaired by incubation with L. johnsonii MG. RNA sequence analysis showed significant upregulation of the expression of genes for tight junctions, anti-inflammatory effects, transcriptional regulation, and apoptosis in Caco-2 cells, following L. johnsonii MG treatment. In L. johnsonii MG, the surface layer 40 kDa protein was purified with gut protein-coupled affinity resin and identified as the moonlighting protein glyceraldehyde-3-phosphate dehydrogenase (GAPDH). These results suggest that L. johnsonii MG promotes the barrier function integrity in Caco-2 cells via GAPDH-JAM-2 binding. Here, we propose a promising approach to identify novel gut receptor molecules based on commensal bacterial interactions and understand host-bacterial communication in a mouse model.
Collapse
Affiliation(s)
- Yuying Bai
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa 226-8501, Japan
| | - Mengying Lyu
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa 226-8501, Japan
| | - Moe Fukunaga
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa 226-8501, Japan
| | - Shin Watanabe
- Department of Emergency and Disaster Medicine Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Shun Iwatani
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa 226-8501, Japan.,Tsukuba Biotechnology Research Center, 5-2-3, Tokodai, Tsukuba-shi, Ibaraki 300-2698, Japan
| | - Kazuhiko Miyanaga
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa 226-8501, Japan.,Department of Infection and Immunity, School of Medicine, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke-Shi, Tochigi, 329-0498, Japan
| | - Naoyuki Yamamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa 226-8501, Japan
| |
Collapse
|
13
|
Bonhomme D, Werts C. Host and Species-Specificities of Pattern Recognition Receptors Upon Infection With Leptospira interrogans. Front Cell Infect Microbiol 2022; 12:932137. [PMID: 35937697 PMCID: PMC9353586 DOI: 10.3389/fcimb.2022.932137] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/24/2022] [Indexed: 12/12/2022] Open
Abstract
Leptospirosis is a zoonotic infectious disease affecting all vertebrates. It is caused by species of the genus Leptospira, among which are the highly pathogenic L. interrogans. Different mammals can be either resistant or susceptible to the disease which can present a large variety of symptoms. Humans are mostly asymptomatic after infection but can have in some cases symptoms varying from a flu-like syndrome to more severe forms such as Weil's disease, potentially leading to multiorgan failure and death. Similarly, cattle, pigs, and horses can suffer from acute forms of the disease, including morbidity, abortion, and uveitis. On the other hand, mice and rats are resistant to leptospirosis despite chronical colonization of the kidneys, excreting leptospires in urine and contributing to the transmission of the bacteria. To this date, the immune mechanisms that determine the severity of the infection and that confer susceptibility to leptospirosis remain enigmatic. To our interest, differential immune sensing of leptospires through the activation of or escape from pattern recognition receptors (PRRs) by microbe-associated molecular patterns (MAMPs) has recently been described. In this review, we will summarize these findings that suggest that in various hosts, leptospires differentially escape recognition by some Toll-like and NOD-like receptors, including TLR4, TLR5, and NOD1, although TLR2 and NLRP3 responses are conserved independently of the host. Overall, we hypothesize that these innate immune mechanisms could play a role in determining host susceptibility to leptospirosis and suggest a central, yet complex, role for TLR4.
Collapse
Affiliation(s)
| | - Catherine Werts
- Institut Pasteur, Université de Paris, CNRS UMR2001, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| |
Collapse
|
14
|
Surdel MC, Anderson PN, Hahn BL, Coburn J. Hematogenous dissemination of pathogenic and non-pathogenic Leptospira in a short-term murine model of infection. Front Cell Infect Microbiol 2022; 12:917962. [PMID: 35923802 PMCID: PMC9339599 DOI: 10.3389/fcimb.2022.917962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/27/2022] [Indexed: 01/19/2023] Open
Abstract
Leptospirosis is an emerging zoonosis caused by pathogenic Leptospira spp. Because rodents are natural hosts of Leptospira, rodent models of pathogenesis have been limited, but are valuable to understand infection in reservoir animals even in the absence of disease. Mouse models of infection provide advantages due to genetic tractability, so developing murine models of Leptospira infection is crucial for further understanding the biology of this organism. Previously our laboratory developed a short-term murine model of Borrelia burgdorferi hematogenous dissemination to investigate the role of adhesion proteins on bacterial survival and dissemination within a host. Here we adapt this model to Leptospira. C3H/HeJ mice are anesthetized, inoculated intravenously, and then bacteria are allowed to circulate for up to twenty-four hours. Mice are euthanized, perfused with saline, and tissues are harvested for culture and DNA purification. Bacterial burdens are determined by quantitative PCR. Reproducible burdens of bacteria were found in tissues upon inoculation with pathogens and non-pathogens, demonstrating the utility of this model to probe different Leptospira species and strains. Pathogenic L. interrogans has a significantly higher burden in blood, liver, kidney, and bladder at one-hour post-inoculation when compared to non-pathogenic L. biflexa. Colonization of the kidney is essential to the life cycle of pathogenic Leptospira in nature. Measurable burdens of non-pathogenic L. biflexa were found in numerous organs and live leptospires were recovered from blood samples for at least three hours post-inoculation, contrary to the previous belief that non-pathogenic leptospires are rapidly cleared. This short-term murine model of Leptospira hematogenous dissemination will allow for the interrogation of virulence factors potentially important for tissue colonization and evasion of host defenses, and represents a novel animal model for investigating determinants of Leptospira infection.
Collapse
Affiliation(s)
- Matthew C. Surdel
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Phillip N. Anderson
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Beth L. Hahn
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jenifer Coburn
- Department of Medicine, Division of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
15
|
Santecchia I, Bonhomme D, Papadopoulos S, Escoll P, Giraud-Gatineau A, Moya-Nilges M, Vernel-Pauillac F, Boneca IG, Werts C. Alive Pathogenic and Saprophytic Leptospires Enter and Exit Human and Mouse Macrophages With No Intracellular Replication. Front Cell Infect Microbiol 2022; 12:936931. [PMID: 35899053 PMCID: PMC9310662 DOI: 10.3389/fcimb.2022.936931] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/14/2022] [Indexed: 02/03/2023] Open
Abstract
Leptospira interrogans are pathogenic bacteria responsible for leptospirosis, a zoonosis impacting 1 million people per year worldwide. Leptospires can infect all vertebrates, but not all hosts develop similar symptoms. Human and cattle may suffer from mild to acute illnesses and are therefore considered as sensitive to leptospirosis. In contrast, mice and rats remain asymptomatic upon infection, although they get chronically colonized in their kidneys. Upon infection, leptospires are stealth pathogens that partially escape the recognition by the host innate immune system. Although leptospires are mainly extracellular bacteria, it was suggested that they could also replicate within macrophages. However, contradictory data in the current literature led us to reevaluate these findings. Using a gentamicin-protection assay coupled to high-content (HC) microscopy, we observed that leptospires were internalized in vivo upon peritoneal infection of C57BL/6J mice. Additionally, three different serotypes of pathogenic L. interrogans and the saprophytic L. biflexa actively infected both human (PMA differentiated) THP1 and mouse RAW264.7 macrophage cell lines. Next, we assessed the intracellular fate of leptospires using bioluminescent strains, and we observed a drastic reduction in the leptospiral intracellular load between 3 h and 6 h post-infection, suggesting that leptospires do not replicate within these cells. Surprisingly, the classical macrophage microbicidal mechanisms (phagocytosis, autophagy, TLR-mediated ROS, and RNS production) were not responsible for the observed decrease. Finally, we demonstrated that the reduction in the intracellular load was associated with an increase of the bacteria in the supernatant, suggesting that leptospires exit both human and murine macrophages. Overall, our study reevaluated the intracellular fate of leptospires and favors an active entrance followed by a rapid exit, suggesting that leptospires do not have an intracellular lifestyle in macrophages.
Collapse
Affiliation(s)
- Ignacio Santecchia
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Delphine Bonhomme
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Stylianos Papadopoulos
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Pedro Escoll
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, Unité Biologie des Bactéries Intracellulaires, Paris, France
| | - Alexandre Giraud-Gatineau
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, Unité de Biologie des Spirochètes, Paris, France
| | - Maryse Moya-Nilges
- Institut Pasteur, Université Cité Paris, Plateforme de Bio-imagerie Ultrastructurale, Paris, France
| | - Frédérique Vernel-Pauillac
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Ivo Gomperts Boneca
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| | - Catherine Werts
- Institut Pasteur, Université Cité Paris, CNRS UMR6047, INSERM U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Paris, France
| |
Collapse
|
16
|
Shu J, Yu H, Ren X, Wang Y, Zhang K, Tang Z, Dang L, Chen W, Li B, Xie H, Li Z. Role of salivary glycopatterns for oral microbiota associated with gastric cancer. Int J Biol Macromol 2022; 209:1368-1378. [PMID: 35461868 DOI: 10.1016/j.ijbiomac.2022.04.133] [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: 12/10/2021] [Revised: 04/01/2022] [Accepted: 04/18/2022] [Indexed: 11/25/2022]
Abstract
Microbiota in the oral cavity plays an important role in maintaining human health. Our previous studies have revealed significant alterations of salivary glycopatterns in gastric cancer (GC) patients, but it is unclear whether these altered salivary glycopatterns can cause the dysbiosis of oral microbiota. In this study, the oral microbiome of healthy volunteers (HVs) and GC patients were detected. The neoglycoproteins were then synthesized according to the altered glycopatterns in GC patients and used to explore the effects of specific salivary glycopattern against oral microbiota. The results showed that five species were significantly increased (p < 0.05) while two species were significantly decreased (p < 0.01) in the saliva of GC patients compared with that of HVs. And the fucose-neoglycoproteins (30-100 μg/mL) could reduce the adhesion and toxicity of Aggregatibacter segnis (A. segnis) to oral cells (HOEC and CAL-27), change the glycan structures of lipopolysaccharide on the surface of A. segnis, and enhance the capacity of A. segnis to trigger innate immune responses. This study revealed that the changes of salivary protein glycopatterns in GC patients might contribute to the dysbiosis of oral microbiota, and had important implications in developing new carbohydrate drugs to maintain a balanced microbiota in the oral.
Collapse
Affiliation(s)
- Jian Shu
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Hanjie Yu
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Xiameng Ren
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Yan Wang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Kun Zhang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Zhen Tang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Liuyi Dang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Wentian Chen
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Baozhen Li
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University Health Science Center, 277 Yanta Xilu, Xi'an 710061, China
| | - Hailong Xie
- Institute of Cancer Research, University of South China, Hengyang, China
| | - Zheng Li
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China.
| |
Collapse
|
17
|
Li J, Ma Y, Paquette JK, Richards AC, Mulvey MA, Zachary JF, Teuscher C, Weis JJ. The Cdkn2a gene product p19 alternative reading frame (p19ARF) is a critical regulator of IFNβ-mediated Lyme arthritis. PLoS Pathog 2022; 18:e1010365. [PMID: 35324997 PMCID: PMC8946740 DOI: 10.1371/journal.ppat.1010365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 02/11/2022] [Indexed: 11/18/2022] Open
Abstract
Type I interferon (IFN) has been identified in patients with Lyme disease, and its abundant expression in joint tissues of C3H mice precedes development of Lyme arthritis. Forward genetics using C3H mice with severe Lyme arthritis and C57BL/6 (B6) mice with mild Lyme arthritis identified the Borrelia burgdorferi arthritis-associated locus 1 (Bbaa1) on chromosome 4 (Chr4) as a regulator of B. burgdorferi-induced IFNβ expression and Lyme arthritis severity. B6 mice introgressed with the C3H allele for Bbaa1 (B6.C3-Bbaa1 mice) displayed increased severity of arthritis, which is initiated by myeloid lineage cells in joints. Using advanced congenic lines, the physical size of the Bbaa1 interval has been reduced to 2 Mbp, allowing for identification of potential genetic regulators. Small interfering RNA (siRNA)-mediated silencing identified Cdkn2a as the gene responsible for Bbaa1 allele-regulated induction of IFNβ and IFN-stimulated genes (ISGs) in bone marrow-derived macrophages (BMDMs). The Cdkn2a-encoded p19 alternative reading frame (p19ARF) protein regulates IFNβ induction in BMDMs as shown by siRNA silencing and overexpression of ARF. In vivo studies demonstrated that p19ARF contributes to joint-specific induction of IFNβ and arthritis severity in B. burgdorferi-infected mice. p19ARF regulates B. burgdorferi-induced IFNβ in BMDMs by stabilizing the tumor suppressor p53 and sequestering the transcriptional repressor BCL6. Our findings link p19ARF regulation of p53 and BCL6 to the severity of IFNβ-induced Lyme arthritis in vivo and indicate potential novel roles for p19ARF, p53, and BCL6 in Lyme disease and other IFN hyperproduction syndromes. Lyme disease is caused by infection with the tick-transmitted bacterium Borrelia burgdorferi. Although different isolates of B. burgdorferi have distinct potential for dissemination and tissue invasion, factors intrinsic to the infected host also play an important role in directing the severity of Lyme disease. In the animal model, infected C3H mice develop severe Lyme arthritis following elevation of type I IFN in joint tissue, while in C57BL/6 (B6) mice arthritis is mild and not associated with type I IFN. We demonstrated that the Borrelia burgdorferi arthritis-associated locus 1 (Bbaa1) on chromosome 4 (Chr4) intrinsically controls the magnitude of IFNβ production and the severity of Lyme arthritis in C3H vs B6 mice. The Cdkn2a gene was positionally identified as the regulator of IFNβ within Bbaa1, and determined to function through its protein product p19 alternative reading frame (p19ARF). ARF regulates IFNβ expression and Lyme arthritis severity by modulating the activities of the tumor suppressor p53 and transcriptional repressor BCL6. Our study provides new insight and potential therapeutic targets for the investigation of type I IFN-dependent Lyme arthritis and other IFN-driven diseases.
Collapse
Affiliation(s)
- Jinze Li
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Ying Ma
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Jackie K. Paquette
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Amanda C. Richards
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Matthew A. Mulvey
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - James F. Zachary
- Department of Veterinary Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Cory Teuscher
- Department of Medicine, Vermont Center for Immunology and Infectious Diseases, Larner College of Medicine, The University of Vermont, Burlington, Vermont, United States of America
| | - Janis J. Weis
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
18
|
Xie X, Lv T, Wu D, Shi H, Zhang S, Xian X, Liu G, Zhang W, Cao Y. IL-10 Deficiency Protects Hamsters from Leptospira Infection. Infect Immun 2022; 90:e0058421. [PMID: 34898251 PMCID: PMC8852706 DOI: 10.1128/iai.00584-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/01/2021] [Indexed: 11/20/2022] Open
Abstract
Leptospirosis is a global zoonotic disease with outcomes ranging from subclinical infection to fatal Weil's syndrome. In addition to antibiotics, some immune activators have shown protective effects against leptospirosis. However, the unclear relationship between Leptospira and cytokines has limited the development of antileptospiral immunomodulators. In this study, the particular role of interleukin-10 (IL-10) in leptospirosis was explored by using IL-10-defective (IL-10-/-) hamsters. After Leptospira infection, an improved survival rate, reduced leptospiral burden, and alleviation of organ lesions were found in IL-10-/- hamsters compared with wild-type (WT) hamsters. In addition, the levels of expression of the IL-1β, IL-6, and tumor necrosis factor alpha (TNF-α) genes and the level of nitric oxide (NO) were higher in IL-10-/- hamsters than in WT hamsters. Our results indicate that IL-10 deficiency protects hamsters from Leptospira infection.
Collapse
Affiliation(s)
- Xufeng Xie
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
| | - Tianbao Lv
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
| | - Dianjun Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
| | - Haozhe Shi
- Institute of Cardiovascular Sciences, Peking University, Beijing, People’s Republic of China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, People’s Republic of China
| | - Shilei Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
| | - Xunde Xian
- Institute of Cardiovascular Sciences, Peking University, Beijing, People’s Republic of China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, People’s Republic of China
| | - George Liu
- Institute of Cardiovascular Sciences, Peking University, Beijing, People’s Republic of China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, People’s Republic of China
| | - Wenlong Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
- Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
| | - Yongguo Cao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
- Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
19
|
Li S, Lei Y, Lei J, Li H. All‑trans retinoic acid promotes macrophage phagocytosis and decreases inflammation via inhibiting CD14/TLR4 in acute lung injury. Mol Med Rep 2021; 24:868. [PMID: 34676874 PMCID: PMC8554390 DOI: 10.3892/mmr.2021.12508] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 02/18/2021] [Indexed: 12/22/2022] Open
Abstract
Acute lung injury (ALI) is a common clinical emergency and all-trans retinoic acid (ATRA) can alleviate organ injury. Therefore, the present study investigated the role of ATRA in ALI. Lipopolysaccharide (LPS)-induced ALI rats were treated with ATRA and the arterial partial pressure of oxygen (PaO2), lung wet/dry weight (W/D) ratio and protein content in the bronchial alveolar lavage fluid (BALF) were measured to evaluate the effect of ATRA on ALI rats. Alveolar macrophages were isolated from the BALF. The phagocytic function of macrophages was detected using the chicken erythrocyte phagocytosis method and flow cytometry. The viability of macrophages was measured using a Cell Counting Kit-8 assay, and apoptosis was analyzed using a TUNEL assay and flow cytometry. The expression levels of Toll-like receptor 4 (TLR4) and cluster of differentiation (CD)14 on the macrophage membrane were detected by immunofluorescence staining. The protein levels of TLR4, CD14, phosphorylated (p)-65, p65, p-IκBα and IκBα were analyzed using western blotting. The concentrations of IL-6, IL-1β and macrophage inflammatory protein-2 in the plasma of rats were detected by ELISA. Macrophages were treated with IAXO-102 (TLR4 inhibitor) to verify the involvement of CD14/TLR4 in the effect of ATRA on ALI. ATRA provided protection against LPS-induced ALI, as evidenced by the increased PaO2 and reduced lung W/D ratio and protein content in the BALF. ATRA enhanced macrophage phagocytosis and viability and reduced apoptosis and inflammation in ALI rats. Mechanically, ATRA inhibited CD14 and TLR4 expression and NF-κB pathway activation. ATRA enhanced macrophage phagocytosis and reduced inflammation by inhibiting the CD14/TLR4-NF-κB pathway in LPS-induced ALI. In summary, ATRA inactivated the NF-κB pathway by inhibiting the expression of CD14/TLR4 receptor in the alveolar macrophages of rats, thus enhancing the phagocytic function of macrophages in ALI rats, improving the activity of macrophages, inhibiting apoptosis, reducing the levels of inflammatory factors, and consequently playing a protective role in ALI model rats. This study may offer novel insights for the clinical management of ALI.
Collapse
Affiliation(s)
- Shuangxue Li
- Department of Respiratory and Critical Care Medicine, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| | - Yuansheng Lei
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jieyun Lei
- Department of Cardiology, Taiyuan Central Hospital, Taiyuan, Shanxi 030009, P.R. China
| | - Hui Li
- Department of Gynecology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, P.R. China
| |
Collapse
|
20
|
Coburn J, Picardeau M, Woods CW, Veldman T, Haake DA. Pathogenesis insights from an ancient and ubiquitous spirochete. PLoS Pathog 2021; 17:e1009836. [PMID: 34673833 PMCID: PMC8530280 DOI: 10.1371/journal.ppat.1009836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Jenifer Coburn
- Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | | | - Christopher W. Woods
- Duke University Medical Center, Durham, North Carolina, United States of America
- Durham VA Medical Center, Durham, North Carolina, United States of America
| | - Timothy Veldman
- Duke University Medical Center, Durham, North Carolina, United States of America
| | - David A. Haake
- VA Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
- The David Geffen School of Medicine at the University of California, Los Angeles, California, United States of America
| |
Collapse
|
21
|
Wang Y, Fan X, Du L, Liu B, Xiao H, Zhang Y, Wu Y, Liu F, Chang YF, Guo X, He P. Scavenger receptor A1 participates in uptake of Leptospira interrogans serovar Autumnalis strain 56606v and inflammation in mouse macrophages. Emerg Microbes Infect 2021; 10:939-953. [PMID: 33929941 PMCID: PMC8153709 DOI: 10.1080/22221751.2021.1925160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Leptospirosis, caused by pathogenic Leptospira species, has emerged as a widespread zoonotic disease worldwide. Macrophages mediate the elimination of pathogens through phagocytosis and cytokine production. Scavenger receptor A1 (SR-A1), one of the critical receptors mediating this process, plays a complicated role in innate immunity. However, the role of SR-A1 in the immune response against pathogenic Leptospira invasion is unknown. In the present study, we found that SR-A1 is an important nonopsonic phagocytic receptor on murine macrophages for Leptospira. However, intraperitoneal injection of leptospires into WT mice presented with more apparent jaundice, subcutaneous hemorrhaging, and higher bacteria burdens in blood and tissues than that of SR-A1-/- mice. Exacerbated cytokine and inflammatory mediator levels were also observed in WT mice and higher recruited macrophages in the liver than those of SR-A1-/- mice. Our findings collectively reveal that although beneficial in the uptake of Leptospira by macrophage, SR-A1 might be exploited by Leptospira to modulate inflammatory activation and increase the susceptibility of infection in the host. These results provide our new insights into the innate immune response during early infection by L. interrogans.
Collapse
Affiliation(s)
- Yanchun Wang
- Department of Medical Microbiology and Parasitology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
| | - Xia Fan
- Department of Medical Microbiology and Parasitology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Lin Du
- Department of Medical Microbiology and Parasitology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Boyu Liu
- Department of Medical Microbiology and Parasitology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Haihan Xiao
- Department of Medical Microbiology and Parasitology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yan Zhang
- Department of Medical Microbiology and Parasitology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yunqiang Wu
- Department of Medical Microbiology and Parasitology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Fuli Liu
- Department of Medical Microbiology and Parasitology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yung-Fu Chang
- College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Xiaokui Guo
- Key Laboratory of Parasite and Vector Biology, Ministry of Health; School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Ping He
- Department of Medical Microbiology and Parasitology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,Key Laboratory of Parasite and Vector Biology, Ministry of Health; School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
22
|
Anti-Leptospira immunoglobulin profiling in mice reveals strain specific IgG and persistent IgM responses associated with virulence and renal colonization. PLoS Negl Trop Dis 2021; 15:e0008970. [PMID: 33705392 PMCID: PMC8007020 DOI: 10.1371/journal.pntd.0008970] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/29/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Leptospira interrogans is a pathogenic spirochete responsible for leptospirosis, a neglected, zoonotic reemerging disease. Humans are sensitive hosts and may develop severe disease. Some animal species, such as rats and mice can become asymptomatic renal carriers. More than 350 leptospiral serovars have been identified, classified on the basis of the antibody response directed against the lipopolysaccharide (LPS). Similarly to whole inactivated bacteria used as human vaccines, this response is believed to confer only short-term, serogroup-specific protection. The immune response of hosts against leptospires has not been thoroughly studied, which complicates the testing of vaccine candidates. In this work, we studied the immunoglobulin (Ig) profiles in mice infected with L. interrogans over time to determine whether this humoral response confers long-term protection after homologous challenge six months post-infection. Groups of mice were injected intraperitoneally with 2×107 leptospires of one of three pathogenic serovars (Manilae, Copenhageni or Icterohaemorrhagiae), attenuated mutants or heat-killed bacteria. Leptospira-specific immunoglobulin (IgA, IgM, IgG and 4 subclasses) produced in the first weeks up to 6 months post-infection were measured by ELISA. Strikingly, we found sustained high levels of IgM in mice infected with the pathogenic Manilae and Copenhageni strains, both colonizing the kidney. In contrast, the Icterohaemorrhagiae strain did not lead to kidney colonization, even at high dose, and triggered a classical IgM response that peaked at day 8 post-infection and disappeared. The virulent Manilae and Copenhageni serovars elicited high levels and similar profiles of IgG subclasses in contrast to Icterohaemorrhagiae strains that stimulated weaker antibody responses. Inactivated heat-killed Manilae strains elicited very low responses. However, all mice pre-injected with leptospires challenged with high doses of homologous bacteria did not develop acute leptospirosis, and all antibody responses were boosted after challenge. Furthermore, we showed that 2 months post-challenge, mice pre-infected with the attenuated M895 Manilae LPS mutant or heat-killed bacterin were completely protected against renal colonization. In conclusion, we observed a sustained IgM response potentially associated with chronic leptospiral renal infection. We also demonstrated in mice different profiles of protective and cross-reactive antibodies after L. interrogans infection, depending on the serovar and virulence of strains. Leptospira interrogans is a pathogenic spirochete responsible for leptospirosis, a neglected zoonotic reemerging disease. The immune response of hosts against these bacteria has not been thoroughly studied. Here, we studied over 6 months the antibody profiles in mice infected with L. interrogans and determined whether this humoral response confers long-term protection after homologous challenge six months after primary infection. Groups of mice were infected intraperitoneally with 2×107 bacteria of one of three different pathogenic serovars (Manilae, Copenhageni and Icterohaemorrhagiae) and some corresponding attenuated avirulent mutants. We measured by ELISA each type of Leptospira-specific immunoglobulin (Ig) (IgA, IgM, IgG and 4 subclasses) produced in the first weeks up to 6 months post-infection and studied their cross-reactivities among serovars. We showed different profiles of antibody response after L. interrogans challenge in mice, depending on the serovar and virulence of strains. However, all infected mice, including the ones harboring low antibody levels, like mice vaccinated with an inactivated, heat-killed strain, were protected against leptospirosis after challenge. Notably, we also showed an unusual sustained IgM response associated with chronic leptospiral colonization. Altogether, this long-term immune protection is different from what is known in humans and warrants further investigation.
Collapse
|
23
|
Nair N, Guedes MS, Hajjar AM, Werts C, Gomes-Solecki M. Role of TLR4 in Persistent Leptospira interrogans Infection: A Comparative In Vivo Study in Mice. Front Immunol 2021; 11:572999. [PMID: 33519799 PMCID: PMC7843520 DOI: 10.3389/fimmu.2020.572999] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022] Open
Abstract
Toll-Like Receptor (TLR) 4, the LPS receptor, plays a central role in the control of leptospirosis and absence of TLR4 results in lethal infection in mice. Because human TLR4 does not sense the atypical leptospiral-LPS, we hypothesized that TLR4/MD-2 humanized transgenic mice (huTLR4) may be more susceptible to leptospirosis than wild-type mice, and thus may constitute a model of acute human leptospirosis. We infected huTLR4 mice, which express human TLR4 but not murine TLR4, with a high dose of L. interrogans serovar Copenhageni FioCruz (Leptospira) in comparison to C57BL/6J wild-type (WT) and, as a control, a congenic strain in which the tlr4 coding sequences are deleted (muTLR4Lps-del). We show that the huTLR4 gene is fully functional in the murine background. We found that dissemination of Leptospira in blood, shedding in urine, colonization of the kidney and overall kinetics of leptospirosis progression is equivalent between WT and huTLR4 C57BL/6J mice. Furthermore, inflammation of the kidney appeared to be subdued in huTLR4 compared to WT mice in that we observed less infiltrates of mononuclear lymphocytes, less innate immune markers and no relevant differences in fibrosis markers. Thus, huTLR4 mice showed less inflammation and kidney pathology, and are not more susceptible to leptospirosis than WT mice. This study is significant as it indicates that one intact TLR4 gene, be it mouse or human, is necessary to control acute leptospirosis.
Collapse
Affiliation(s)
- Nisha Nair
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Mariana S. Guedes
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Adeline M. Hajjar
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Catherine Werts
- Institut Pasteur, Biology and Genetics of the Bacterial Cell Wall Unit, Paris, France,CNRS, UMR 2001 Microbiologie intégrative et Moléculaire, Paris, France
| | - Maria Gomes-Solecki
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States,*Correspondence: Maria Gomes-Solecki,
| |
Collapse
|
24
|
Bonhomme D, Santecchia I, Vernel-Pauillac F, Caroff M, Germon P, Murray G, Adler B, Boneca IG, Werts C. Correction: Leptospiral LPS escapes mouse TLR4 internalization and TRIF-associated antimicrobial responses through O antigen and associated lipoproteins. PLoS Pathog 2020; 16:e1009173. [PMID: 33362240 PMCID: PMC7757799 DOI: 10.1371/journal.ppat.1009173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
25
|
Santecchia I, Ferrer MF, Vieira ML, Gómez RM, Werts C. Phagocyte Escape of Leptospira: The Role of TLRs and NLRs. Front Immunol 2020; 11:571816. [PMID: 33123147 PMCID: PMC7573490 DOI: 10.3389/fimmu.2020.571816] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022] Open
Abstract
The spirochetal bacteria Leptospira spp. are causative agents of leptospirosis, a globally neglected and reemerging zoonotic disease. Infection with these pathogens may lead to an acute and potentially fatal disease but also to chronic asymptomatic renal colonization. Both forms of disease demonstrate the ability of leptospires to evade the immune response of their hosts. In this review, we aim first to recapitulate the knowledge and explore the controversial data about the opsonization, recognition, intracellular survival, and killing of leptospires by scavenger cells, including platelets, neutrophils, macrophages, and dendritic cells. Second, we will summarize the known specificities of the recognition or escape of leptospire components (the so-called microbial-associated molecular patterns; MAMPs) by the pattern recognition receptors (PRRs) of the Toll-like and NOD-like families. These PRRs are expressed by phagocytes, and their stimulation by MAMPs triggers pro-inflammatory cytokine and chemokine production and bactericidal responses, such as antimicrobial peptide secretion and reactive oxygen species production. Finally, we will highlight recent studies suggesting that boosting or restoring phagocytic functions by treatments using agonists of the Toll-like or NOD receptors represents a novel prophylactic strategy and describe other potential therapeutic or vaccine strategies to combat leptospirosis.
Collapse
Affiliation(s)
- Ignacio Santecchia
- Institut Pasteur, Microbiology Department, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- CNRS, UMR 2001 Microbiologie intégrative et Moléculaire, Paris, France
- INSERM, Equipe Avenir, Paris, France
- Université de Paris, Sorbonne Paris Cité, Paris, France
| | - María Florencia Ferrer
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - Monica Larucci Vieira
- Departamento de Microbiologia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ricardo Martín Gómez
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - Catherine Werts
- Institut Pasteur, Microbiology Department, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- CNRS, UMR 2001 Microbiologie intégrative et Moléculaire, Paris, France
| |
Collapse
|
26
|
Kędzierska-Mieszkowska S, Arent Z. AAA+ Molecular Chaperone ClpB in Leptospira interrogans: Its Role and Significance in Leptospiral Virulence and Pathogenesis of Leptospirosis. Int J Mol Sci 2020; 21:E6645. [PMID: 32932775 PMCID: PMC7555560 DOI: 10.3390/ijms21186645] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/31/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
Bacterial ClpB is an ATP-dependent disaggregase that belongs to the Hsp100/Clp subfamily of the AAA+ ATPases and cooperates with the DnaK chaperone system in the reactivation of aggregated proteins, as well as promotes bacterial survival under adverse environmental conditions, including thermal and oxidative stresses. In addition, extensive evidence indicates that ClpB supports the virulence of numerous bacteria, including pathogenic spirochaete Leptospira interrogans responsible for leptospirosis in animals and humans. However, the specific function of ClpB in leptospiral virulence still remains to be fully elucidated. Interestingly, ClpB was predicted as one of the L. interrogans hub proteins interacting with human proteins, and pathogen-host protein interactions are fundamental for successful invasion of the host immune system by bacteria. The aim of this review is to discuss the most important aspects of ClpB's function in L. interrogans, including contribution of ClpB to leptospiral virulence and pathogenesis of leptospirosis, a zoonotic disease with a significant impact on public health worldwide.
Collapse
Affiliation(s)
| | - Zbigniew Arent
- University Centre of Veterinary Medicine, University of Agriculture in Krakow, 30-059 Krakow, Poland;
| |
Collapse
|
27
|
Holzapfel M, Bonhomme D, Cagliero J, Vernel-Pauillac F, Fanton d’Andon M, Bortolussi S, Fiette L, Goarant C, Wunder EA, Picardeau M, Ko AI, Werling D, Matsui M, Boneca IG, Werts C. Escape of TLR5 Recognition by Leptospira spp.: A Rationale for Atypical Endoflagella. Front Immunol 2020; 11:2007. [PMID: 32849665 PMCID: PMC7431986 DOI: 10.3389/fimmu.2020.02007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/24/2020] [Indexed: 12/26/2022] Open
Abstract
Leptospira (L.) interrogans are invasive bacteria responsible for leptospirosis, a worldwide zoonosis. They possess two periplasmic endoflagellae that allow their motility. L. interrogans are stealth pathogens that escape the innate immune recognition of the NOD-like receptors NOD1/2, and the human Toll-like receptor (TLR)4, which senses peptidoglycan and lipopolysaccharide (LPS), respectively. TLR5 is another receptor of bacterial cell wall components, recognizing flagellin subunits. To study the contribution of TLR5 in the host defense against leptospires, we infected WT and TLR5 deficient mice with pathogenic L. interrogans and tracked the infection by in vivo live imaging of bioluminescent bacteria or by qPCR. We did not identify any protective or inflammatory role of murine TLR5 for controlling pathogenic Leptospira. Likewise, subsequent in vitro experiments showed that infections with different live strains of L. interrogans and L. biflexa did not trigger TLR5 signaling. However, unexpectedly, heat-killed bacteria stimulated human and bovine TLR5, but did not, or barely induced stimulation via murine TLR5. Abolition of TLR5 recognition required extensive boiling time of the bacteria or proteinase K treatment, showing an unusual high stability of the leptospiral flagellins. Interestingly, after using antimicrobial peptides to destabilize live leptospires, we detected TLR5 activity, suggesting that TLR5 could participate in the fight against leptospires in humans or cattle. Using different Leptospira strains with mutations in the flagellin proteins, we further showed that neither FlaA nor Fcp participated in the recognition by TLR5, suggesting a role for the FlaB. FlaB have structural homology to Salmonella FliC, and possess conserved residues important for TLR5 activation, as shown by in silico analyses. Accordingly, we found that leptospires regulate the expression of FlaB mRNA according to the growth phase in vitro, and that infection with L. interrogans in hamsters and in mice downregulated the expression of the FlaB, but not the FlaA subunits. Altogether, in contrast to different bacteria that modify their flagellin sequences to escape TLR5 recognition, our study suggests that the peculiar central localization and stability of the FlaB monomers in the periplasmic endoflagellae, associated with the downregulation of FlaB subunits in hosts, constitute an efficient strategy of leptospires to escape the TLR5 recognition and the induced immune response.
Collapse
Affiliation(s)
- Marion Holzapfel
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- CNRS, UMR 2001 Microbiologie Intégrative et Moléculaire, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Equipe Avenir, Paris, France
| | - Delphine Bonhomme
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- CNRS, UMR 2001 Microbiologie Intégrative et Moléculaire, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Equipe Avenir, Paris, France
- Sorbonne Paris Cité, Université de Paris, Paris, France
| | - Julie Cagliero
- Institut Pasteur de Nouvelle Calédonie, Immunity and Inflammation Group, Institut Pasteur International Network, Noumea, France
| | - Frédérique Vernel-Pauillac
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- CNRS, UMR 2001 Microbiologie Intégrative et Moléculaire, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Equipe Avenir, Paris, France
| | - Martine Fanton d’Andon
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- CNRS, UMR 2001 Microbiologie Intégrative et Moléculaire, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Equipe Avenir, Paris, France
| | - Sophia Bortolussi
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- CNRS, UMR 2001 Microbiologie Intégrative et Moléculaire, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Equipe Avenir, Paris, France
| | - Laurence Fiette
- Unité Histopathologie Humaine et Modèles Animaux, Institut Pasteur, Paris, France
| | - Cyrille Goarant
- Leptospirosis Research and Expertise Unit, Institut Pasteur International Network, Institut Pasteur de Nouvelle Calédonie, Noumea, France
| | - Elsio A. Wunder
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Brazilian Ministry of Health, Salvador, Brazil
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, United States
| | | | - Albert I. Ko
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Brazilian Ministry of Health, Salvador, Brazil
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, United States
| | - Dirk Werling
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hatfield, United Kingdom
| | - Mariko Matsui
- Institut Pasteur de Nouvelle Calédonie, Immunity and Inflammation Group, Institut Pasteur International Network, Noumea, France
| | - Ivo G. Boneca
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- CNRS, UMR 2001 Microbiologie Intégrative et Moléculaire, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Equipe Avenir, Paris, France
| | - Catherine Werts
- Institut Pasteur, Unité Biologie et Génétique de la Paroi Bactérienne, Paris, France
- CNRS, UMR 2001 Microbiologie Intégrative et Moléculaire, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Equipe Avenir, Paris, France
| |
Collapse
|