Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Proc Natl Acad Sci U S A ; 121(22): e2310864121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38781213

ABSTRACT

IL-22 plays a critical role in defending against mucosal infections, but how IL-22 production is regulated is incompletely understood. Here, we show that mice lacking IL-33 or its receptor ST2 (IL-1RL1) were more resistant to Streptococcus pneumoniae lung infection than wild-type animals and that single-nucleotide polymorphisms in IL33 and IL1RL1 were associated with pneumococcal pneumonia in humans. The effect of IL-33 on S. pneumoniae infection was mediated by negative regulation of IL-22 production in innate lymphoid cells (ILCs) but independent of ILC2s as well as IL-4 and IL-13 signaling. Moreover, IL-33's influence on IL-22-dependent antibacterial defense was dependent on housing conditions of the mice and mediated by IL-33's modulatory effect on the gut microbiota. Collectively, we provide insight into the bidirectional crosstalk between the innate immune system and the microbiota. We conclude that both genetic and environmental factors influence the gut microbiota, thereby impacting the efficacy of antibacterial immune defense and susceptibility to pneumonia.


Subject(s)
Immunity, Innate , Interleukin-1 Receptor-Like 1 Protein , Interleukin-22 , Interleukin-33 , Interleukins , Streptococcus pneumoniae , Animals , Interleukin-33/immunology , Interleukin-33/genetics , Interleukin-33/metabolism , Interleukins/metabolism , Interleukins/immunology , Interleukins/genetics , Mice , Streptococcus pneumoniae/immunology , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/immunology , Humans , Mice, Knockout , Microbiota/immunology , Mice, Inbred C57BL , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/microbiology , Gastrointestinal Microbiome/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Polymorphism, Single Nucleotide
2.
Parasitol Res ; 120(1): 209-221, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33263166

ABSTRACT

Schistosomiasis is one of the most devastating parasitic disease in the world. Schistosoma spp. survive for decades within the vasculature of their human hosts. They have evolved a vast array of mechanisms to avoid the immune reaction of the host. Due to their sexual dimorphism, with the female worm lying within the gynecophoric canal of the male worm, it is the male that is exposed to the immediate environment and the soluble parts of the host's immune response. To understand how the worms are so successful in fending off the immune attacks of the host, comparative analyses of both worm sexes in human serum (with or without Praziquantel) were performed using scanning electron microscopy, transmission electron microscopy, and immunohistochemistry. Further, gene expression analyses of tegument-specific genes were performed. Following the incubation in human serum, males and females out of pairs show morphological changes such as an altered structure of the pits below the surface and an increased number of pits per area. In addition, female schistosomes presented a marked tuft-like repulsion of their opsonized surface. The observed resistance of females to Praziquantel seemed to depend on active proteins in the human serum. Moreover, different expression profiles of tegument-specific genes indicate different functions of female_single and male_single teguments in response to human serum. Our results indicate that female schistosomes developed different evasion strategies toward the host's immune system in comparison to males that might lead to more robustness and has to be taken into account for the development of new anti-schistosomal drugs.


Subject(s)
Anthelmintics/pharmacology , Helminth Proteins/metabolism , Praziquantel/pharmacology , Schistosoma/drug effects , Serum/physiology , Animals , Drug Resistance , Female , Helminth Proteins/genetics , Humans , Immune Evasion , Male , Schistosoma/metabolism , Schistosoma/ultrastructure , Sex Factors
3.
Gesundheitswesen ; 83(11): 890-893, 2021 Nov.
Article in German | MEDLINE | ID: mdl-34571553

ABSTRACT

BACKGROUND: The current risk of infection with SARS-CoV-2 in schools continues to be a subject of controversy. METHODOLOGY: "schugi-MV" collects data on the incidence of infection, hygiene management and other factors in structured inspections of schools in Mecklenburg-Western Pomerania. Recommendations for safe teaching are to be derived from the results. This article presents information on the first 10 schools visited between 18.12.2020 and 20.01.2021. RESULTS: At the schools visited, the ratio of the number of index cases among adults and children was 1:1.25. The inspections showed a great heterogeneity of schools and school buildings and the resulting possibilities for implementing infection control measures. CONCLUSION: Based on the present preliminary results, hygiene and infection control measures at schools in Mecklenburg-Western Pomerania cannot be standardised, but should leave room for design.


Subject(s)
COVID-19 , Adult , Child , Germany/epidemiology , Humans , Pandemics/prevention & control , SARS-CoV-2 , Schools
4.
Gastroenterology ; 157(3): 777-792.e14, 2019 09.
Article in English | MEDLINE | ID: mdl-31078624

ABSTRACT

BACKGROUND & AIMS: We studied the role of interleukin 11 (IL11) signaling in the pathogenesis of nonalcoholic steatohepatitis (NASH) using hepatic stellate cells (HSCs), hepatocytes, and mouse models of NASH. METHODS: We stimulated mouse and human fibroblasts, HSCs, or hepatocytes with IL11 and other cytokines and analyzed them by imaging, immunoblot, and functional assays and enzyme-linked immunosorbent assays. Mice were given injections of IL11. Mice with disruption of the interleukin 11 receptor subunit alpha1 gene (Il11ra1-/-) mice and Il11ra1+/+ mice were fed a high-fat methionine- and choline-deficient diet (HFMCD) or a Western diet with liquid fructose (WDF) to induce steatohepatitis; control mice were fed normal chow. db/db mice were fed with methionine- and choline-deficient diet for 12 weeks and C57BL/6 NTac were fed with HFMCD for 10 weeks or WDF for 16 weeks. Some mice were given intraperitoneal injections of anti-IL11 (X203), anti-IL11RA (X209), or a control antibody at different timepoints on the diets. Livers and blood were collected; blood samples were analyzed by biochemistry and liver tissues were analyzed by histology, RNA sequencing, immunoblots, immunohistochemistry, hydroxyproline, and mass cytometry time of flight assays. RESULTS: HSCs incubated with cytokines produced IL11, resulting in activation (phosphorylation) of ERK and expression of markers of fibrosis. Livers of mice given injections of IL11 became damaged, with increased markers of fibrosis, hepatocyte cell death and inflammation. Following the HFMCD or WDF, livers from Il11ra1-/- mice had reduced steatosis, fibrosis, expression of markers of inflammation and steatohepatitis, compared to and Il11ra1+/+ mice on the same diets. Depending on the time of administration of anti-IL11 or anti-IL11RA antibodies to wild-type mice on the HFMCD or WDF, or to db/db mice on the methionine and choline-deficient diet, the antibodies prevented, stopped, or reversed development of fibrosis and steatosis. Blood samples from Il11ra1+/+ mice fed the WDF and given injections of anti-IL11 or anti-IL11RA, as well as from Il11ra1-/- mice fed WDF, had lower serum levels of lipids and glucose than mice not injected with antibody or with disruption of Il11ra1. CONCLUSIONS: Neutralizing antibodies that block IL11 signaling reduce fibrosis, steatosis, hepatocyte death, inflammation and hyperglycemia in mice with diet-induced steatohepatitis. These antibodies also improve the cardiometabolic profile of mice and might be developed for the treatment of NASH.


Subject(s)
Antibodies, Neutralizing/pharmacology , Hepatitis/prevention & control , Interleukin-11 Receptor alpha Subunit/metabolism , Interleukin-11/antagonists & inhibitors , Liver Cirrhosis, Experimental/prevention & control , Liver/drug effects , Non-alcoholic Fatty Liver Disease/prevention & control , Animals , Cell Death/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatitis/genetics , Hepatitis/metabolism , Hepatitis/pathology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inflammation Mediators/metabolism , Interleukin-11/metabolism , Interleukin-11 Receptor alpha Subunit/deficiency , Interleukin-11 Receptor alpha Subunit/genetics , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction/drug effects , THP-1 Cells
5.
Curr Top Microbiol Immunol ; 397: 215-27, 2016.
Article in English | MEDLINE | ID: mdl-27460812

ABSTRACT

Streptococcus pneumoniae frequently colonizes the upper respiratory tract of healthy individuals, but also commonly causes severe invasive infections such as community-acquired pneumonia and meningitis. One of the key virulence factors of pneumococci is the pore-forming toxin pneumolysin which stimulates cell death and is involved in the evasion of some defense mechanisms. The immune system, however, employs different inflammasomes to sense pneumolysin-induced pore formation, cellular membrane damage, and/or subsequent leakage of bacterial nucleic acid into the host cell cytosol. Canonical inflammasomes are cytosolic multiprotein complexes consisting of a receptor molecule such as NLRP3 or AIM2, the adapter ASC, and caspase-1. NLRP3 and AIM2 inflammasomes mediate cell death and production of important IL-1 family cytokines to recruit leukocytes and defend against S. pneumoniae. Here, we review recent evidence that highlights inflammasomes as critical sensors of S. pneumoniae-induced cellular perturbations, summarize their role in pneumococcal infections, and discuss potential evasion strategies of some emerging pneumococcal strains.


Subject(s)
Immune Evasion , Immunity, Innate , Inflammasomes/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , Animals , Humans , Inflammasomes/genetics , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/physiology
6.
Front Cell Infect Microbiol ; 12: 893632, 2022.
Article in English | MEDLINE | ID: mdl-35865813

ABSTRACT

Background: Schistosomiasis is a severe parasitic disease that is primarily driven by the host's immune response to schistosome eggs trapped in tissue and by the granulomatous inflammatory and fibrotic reaction they cause. Despite significant progress in understanding the complex immunological processes involved in the relationship between schistosomes and their host, neither an effective vaccine against the infection nor anti-fibrotic drugs currently exists, making the search for new targets for schistosome drugs and vaccine candidates even more important. In order to identify new molecular targets for defense against or elimination of the parasite, we investigate herein the interplay between the host and male or female schistosomes, clearly separating this from the action of the parasite eggs. Methods: For this purpose, we infected 6-8-week-old female NMRI mice with 100 male (M), female (F), or both (MF) S. mansoni cercariae and performed a comparative transcriptomic and flow cytometric analysis of their spleens. Results: Principal component analysis of a total of 22,207 transcripts showed a clear clustering of the experimental groups. We identified a total of 1,293 genes in group M, 512 genes in group F, and 4,062 genes in group MF that were differentially expressed compared to naive controls. The highest percentage of regulated genes (2,972; 65.9%) was found in group MF alone, but there was a large overlap between groups M and MF (798; 17.7%) and a small overlap between groups F and MF (91; 2.0%). Only 4.5% of genes (201) were revealed to be regulated in all experimental groups (M/F/MF). In addition, we were able to show that both worm sexes trigger immune responses in an egg-independent manner (non-polarized Th1 and Th2 response), with female worms exerting less regulatory influence than males. Conclusion: Our data show that adult schistosomes trigger sex-specific, egg-independent immune responses. The lists of genes regulated by adult female or male worms presented here may be useful in deciphering host-parasite interactions to identify targets for schistosome elimination.


Subject(s)
Schistosomiasis mansoni , Schistosomiasis , Animals , Female , Male , Mice , Schistosoma mansoni/genetics , Spleen , Transcriptome
7.
Front Immunol ; 13: 1010932, 2022.
Article in English | MEDLINE | ID: mdl-36505463

ABSTRACT

In exhibiting gonochorism and phenotypic sexual dimorphism, Schistosoma spp. are unique among trematodes. Only females mating with male schistosomes can produce the highly immunogenic parasite eggs which determine the clinical picture of the disease schistosomiasis. The strong immune-modulatory effect of the eggs masks the influence of the adult worms. To shed light on the complexity of the immune response triggered by adult worms of Schistosoma mansoni, we performed a long-term unisexual infection experiment in mice. We were able to demonstrate that both male and female schistosomes can survive unpaired for one year in the murine host. Furthermore, unisexual S. mansoni infection leads to pronounced inflammation of the liver characterized by a non-polarized Th1/Th2 immune response, regardless of worm sex.


Subject(s)
Schistosomiasis mansoni , Female , Male , Mice , Animals , T-Lymphocytes , Causality , Schistosoma mansoni , Cell Communication
8.
Front Bioeng Biotechnol ; 9: 686192, 2021.
Article in English | MEDLINE | ID: mdl-34249887

ABSTRACT

In recent decades, biofilm-associated infections have become a major problem in many medical fields, leading to a high burden on patients and enormous costs for the healthcare system. Microbial infestations are caused by opportunistic pathogens which often enter the incision already during implantation. In the subsequently formed biofilm bacteria are protected from the hosts immune system and antibiotic action. Therefore, the development of modified, anti-microbial implant materials displays an indispensable task. Thermoplastic polyurethane (TPU) represents the state-of-the-art material in implant manufacturing. Due to the constantly growing areas of application and the associated necessary adjustments, the optimization of these materials is essential. In the present study, modified liquid silicone rubber (LSR) surfaces were compared with two of the most commonly used TPUs in terms of bacterial colonization and biofilm formation. The tests were conducted with the clinically relevant bacterial strains Staphylococcus aureus and Staphylococcus epidermidis. Crystal violet staining and scanning electron microscopy showed reduced adhesion of bacteria and thus biofilm formation on these new materials, suggesting that the investigated materials are promising candidates for implant manufacturing.

9.
Protoplasma ; 257(5): 1277-1287, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32462473

ABSTRACT

As part of the parasite's excretory/secretory system, extracellular vesicles (EVs) represent a potent communication tool of schistosomes with their human host to strike the balance between their own survival in a hostile immunological environment and a minimal damage to the host tissue. Their cargo consists of functional proteins, lipids, and nucleic acids that facilitate biological processes like migration, nutrient acquisition, or reproduction. The most important impact of the vesicle-mediated communication, however, is the promotion of the parasite survival via mimicking host protein function and directly or indirectly modulating the immune response of the host. Overcoming this shield of immunological adaption in the schistosome-host relation is the aim of current research activities in this field and crucial for the development of a reliable anti-schistosomal therapy. Not least because of their prospective use in clinical applications, research on EVs is now a rapidly expanding field. We herein focus on the current state of knowledge of vesicle-based communication of schistosomes and discussing the role of EVs in facilitating biological processes and immune modulatory properties of EVs considering the different life stages of the parasite.


Subject(s)
Extracellular Vesicles/metabolism , Host-Pathogen Interactions/genetics , Parasites/pathogenicity , Schistosoma/metabolism , Animals , Humans
10.
Biomed Res Int ; 2019: 1704238, 2019.
Article in English | MEDLINE | ID: mdl-31950032

ABSTRACT

BACKGROUND: Hepatic fibrosis and granuloma formation as a consequence of tissue entrapped eggs produced by female schistosomes characterize the pathology of Schistosoma mansoni infection. We have previously shown that single-sex infection with female schistosomes mitigates hepatic fibrosis after secondary infection. This was associated with an increased expression of cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), known as a negative regulator of T cell activation. Based on these findings, we hypothesized that administration of agonistic CTLA-4-Ig (Belatacept) is capable to prevent and/or treat hepatic fibrosis during schistosomiasis. METHODS: Mice were infected with 50 S. mansoni cercariae and CTLA-4-Ig, or appropriated control-Ig was administered for 4 weeks. Preventive treatment started 4 weeks after infection, before onset of egg production, and therapeutic treatment started 8 weeks after infection when hepatic fibrosis was already established. RESULTS: When given early after infection, livers of CTLA-4-Ig-treated mice showed significantly reduced collagen deposition and decreased expression of profibrotic genes in comparison to controls. In addition, administration of CTLA-4-Ig suppressed the inflammatory T cell response in infected mice. If therapy was started at a later time point when fibrogenesis was initiated, CTLA-4-Ig had no impact on hepatic fibrosis. CONCLUSION: We could demonstrate that an early preventive administration of CTLA-4-Ig suppresses effector T cell function and therefore ameliorates liver fibrosis. CTLA-4-Ig administration after onset of egg production fails to treat hepatic fibrosis.


Subject(s)
CTLA-4 Antigen/genetics , Liver Cirrhosis/prevention & control , Schistosomiasis mansoni/drug therapy , T-Lymphocytes/drug effects , Abatacept/pharmacology , Animals , CTLA-4 Antigen/agonists , Disease Models, Animal , Flow Cytometry , Humans , Liver/drug effects , Liver/parasitology , Liver Cirrhosis/immunology , Liver Cirrhosis/parasitology , Liver Cirrhosis/pathology , Mice , Schistosoma mansoni/drug effects , Schistosoma mansoni/pathogenicity , Schistosomiasis mansoni/parasitology , Schistosomiasis mansoni/pathology , T-Lymphocytes/parasitology
11.
Front Immunol ; 9: 861, 2018.
Article in English | MEDLINE | ID: mdl-29743881

ABSTRACT

Infection with the intravascular diecious trematode Schistosoma spp. remains a serious tropical disease and public health problem in the developing world, affecting over 258 million people worldwide. During chronic Schistosoma mansoni infection, complex immune responses to tissue-entrapped parasite eggs provoke granulomatous inflammation which leads to serious damage of the liver and intestine. The suppression of protective host immune mechanisms by helminths promotes parasite survival and benefits the host by reducing tissue damage. However, immune-suppressive cytokines may reduce vaccine-induced immune responses. By combining a single-sex infection system with a murine air pouch model, we were able to demonstrate that male and female schistosomes play opposing roles in modulating the host's immune response. Female schistosomes suppress early innate immune responses to invading cercariae in the skin and upregulate anergy-associated genes. In contrast, male schistosomes trigger strong innate immune reactions which lead to a reduction in worm and egg burden in the liver. Our data suggest that the female worm is a neglected player in the dampening of the host's immune defense system and is therefore a promising target for new immune modulatory therapies.


Subject(s)
Cercaria/immunology , Host-Parasite Interactions/immunology , Protozoan Vaccines/therapeutic use , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Animals , Antibodies, Helminth/immunology , Biomphalaria/parasitology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Immune Tolerance/immunology , Liver/immunology , Liver/parasitology , Male , Mice , Protozoan Vaccines/immunology , Schistosomiasis mansoni/parasitology , Schistosomiasis mansoni/prevention & control , Sex Factors
12.
PLoS One ; 10(2): e0117022, 2015.
Article in English | MEDLINE | ID: mdl-25658823

ABSTRACT

The innate immune system employs C-type lectin receptors (CLRs) to recognize carbohydrate structures on pathogens and self-antigens. The Macrophage-inducible C-type lectin (Mincle) is a FcRγ-coupled CLR that was shown to bind to mycobacterial cord factor as well as certain fungal species. However, since CLR functions during bacterial infections have not yet been investigated thoroughly, we aimed to examine their function in Streptococcus pneumonia infection. Binding studies using a library of recombinantly expressed CLR-Fc fusion proteins indicated a specific, Ca2+-dependent, and serotype-specific binding of Mincle to S. pneumonia. Subsequent experiments with different Mincle-expressing cells as well as Mincle-deficient mice, however, revealed a limited role of this receptor in bacterial phagocytosis, neutrophil-mediated killing, cytokine production, and antibacterial immune response during pneumonia. Collectively, our results indicate that Mincle is able to recognize S. pneumonia but is not required for the anti-pneumococcal innate immune response.


Subject(s)
Immunity, Innate , Lectins, C-Type/metabolism , Membrane Proteins/metabolism , Streptococcus pneumoniae/metabolism , Animals , Calcium/metabolism , Cells, Immobilized/chemistry , Cells, Immobilized/metabolism , Cytokines/analysis , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Phagocytosis , Pneumonia/pathology , Pneumonia/veterinary , Protein Binding , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Streptococcus pneumoniae/chemistry
13.
PLoS One ; 10(8): e0137108, 2015.
Article in English | MEDLINE | ID: mdl-26317436

ABSTRACT

Streptococcus pneumoniae is a major cause of pneumonia, sepsis and meningitis. The pore-forming toxin pneumolysin is a key virulence factor of S. pneumoniae, which can be sensed by the NLRP3 inflammasome. Among the over 90 serotypes, serotype 1 pneumococci (particularly MLST306) have emerged across the globe as a major cause of invasive disease. The cause for its particularity is, however, incompletely understood. We therefore examined pneumococcal infection in human cells and a human lung organ culture system mimicking infection of the lower respiratory tract. We demonstrate that different pneumococcal serotypes differentially activate inflammasome-dependent IL-1ß production in human lung tissue and cells. Whereas serotype 2, 3, 6B, 9N pneumococci expressing fully haemolytic pneumolysins activate NLRP3 inflammasome-dependent responses, serotype 1 and 8 strains expressing non-haemolytic toxins are poor activators of IL-1ß production. Accordingly, purified haemolytic pneumolysin but not serotype 1-associated non-haemolytic toxin activates strong IL-1ß production in human lungs. Our data suggest that the evasion of inflammasome-dependent innate immune responses by serotype 1 pneumococci might contribute to their ability to cause invasive diseases in humans.


Subject(s)
Inflammasomes/metabolism , Lung/metabolism , Lung/microbiology , Streptococcus pneumoniae/physiology , Bacterial Proteins/metabolism , Caspase 1/metabolism , Hemolysis , Humans , Interleukin-1beta/biosynthesis , Lung/cytology , Lung/immunology , Species Specificity , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/metabolism , Streptolysins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL