Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
Nat Microbiol ; 9(7): 1792-1811, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38862602

ABSTRACT

The Klebsiella oxytoca species complex is part of the human microbiome, especially during infancy and childhood. K. oxytoca species complex strains can produce enterotoxins, namely, tilimycin and tilivalline, while also contributing to colonization resistance (CR). The relationship between these seemingly contradictory roles is not well understood. Here, by coupling ex vivo assays with CRISPR-mutagenesis and various mouse models, we show that K. oxytoca provides CR against Salmonella Typhimurium. In vitro, the antimicrobial activity against various Salmonella strains depended on tilimycin production and was induced by various simple carbohydrates. In vivo, CR against Salmonella depended on toxin production in germ-free mice, while it was largely toxin-independent in mice with residual microbiota. This was linked to the relative levels of toxin-inducing carbohydrates in vivo. Finally, dulcitol utilization was essential for toxin-independent CR in gnotobiotic mice. Together, this demonstrates that nutrient availability is key to both toxin-dependent and substrate-driven competition between K. oxytoca and Salmonella.


Subject(s)
Klebsiella oxytoca , Salmonella Infections , Salmonella typhimurium , Klebsiella oxytoca/genetics , Klebsiella oxytoca/metabolism , Animals , Mice , Salmonella Infections/microbiology , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Salmonella typhimurium/growth & development , Salmonella typhimurium/drug effects , Humans , Disease Models, Animal , Enterotoxins/metabolism , Enterotoxins/genetics , Female , Mice, Inbred C57BL , Klebsiella Infections/microbiology , Microbiota , Gastrointestinal Microbiome , Antibiosis , Benzodiazepinones
2.
Eur J Immunol ; 53(10): e2250270, 2023 10.
Article in English | MEDLINE | ID: mdl-37366299

ABSTRACT

Mucosal barrier integrity and pathogen clearance is a complex process influenced by both Th17 and Treg cells. Previously, we had described the DNA methylation profile of Th17 cells and identified Zinc finger protein (Zfp)362 to be uniquely demethylated. Here, we generated Zfp362-/- mice to unravel the role of Zfp362 for Th17 cell biology. Zfp362-/- mice appeared clinically normal, showed no phenotypic alterations in the T-cell compartment, and upon colonization with segmented filamentous bacteria, no effect of Zfp362 deficiency on Th17 cell differentiation was observed. By contrast, Zfp362 deletion resulted in increased frequencies of colonic Foxp3+ Treg cells and IL-10+ and RORγt+ Treg cell subsets in mesenteric lymph nodes. Adoptive transfer of naïve CD4+ T cells from Zfp362-/- mice into Rag2-/- mice resulted in a significantly lower weight loss when compared with controls receiving cells from Zfp362+/+ littermates. However, this attenuated weight loss did not correlate with alterations of Th17 cells but instead was associated with an increase of effector Treg cells in mesenteric lymph nodes. Together, these results suggest that Zfp362 plays an important role in promoting colonic inflammation; however, this function is derived from constraining the effector function of Treg cells rather than directly promoting Th17 cell differentiation.


Subject(s)
T-Lymphocytes, Regulatory , Th17 Cells , Mice , Animals , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Cell Differentiation , Inflammation/metabolism , Weight Loss , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism
3.
Cell Rep ; 42(6): 112549, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37245209

ABSTRACT

Transfer of the gut microbiota from wild to laboratory mice alters the host's immune status and enhances resistance to infectious and metabolic diseases, but understanding of which microbes and how they promote host fitness is only emerging. Our analysis of metagenomic sequencing data reveals that Helicobacter spp. are enriched in wild compared with specific-pathogen-free (SPF) and conventionally housed mice, with multiple species commonly co-colonizing their hosts. We create laboratory mice harboring three non-SPF Helicobacter spp. to evaluate their effect on mucosal immunity and colonization resistance to the enteropathogen Citrobacter rodentium. Our experiments reveal that Helicobacter spp. interfere with C. rodentium colonization and attenuate C. rodentium-induced gut inflammation in wild-type (WT) mice, even preventing lethal infection in Rag2-/- SPF mice. Further analyses suggest that Helicobacter spp. interfere with tissue attachment of C. rodentium, putatively by reducing the availability of mucus-derived sugars. These results unveil pivotal protective functions of wild mouse microbiota constituents against intestinal infection.


Subject(s)
Enterobacteriaceae Infections , Gastrointestinal Microbiome , Microbiota , Animals , Mice , Citrobacter rodentium , Adaptive Immunity , Mice, Inbred C57BL
4.
Viruses ; 15(2)2023 01 18.
Article in English | MEDLINE | ID: mdl-36851486

ABSTRACT

The COVID-19 pandemic remains a global health threat and novel antiviral strategies are urgently needed. SARS-CoV-2 employs the cellular serine protease TMPRSS2 for entry into lung cells, and TMPRSS2 inhibitors are being developed for COVID-19 therapy. However, the SARS-CoV-2 Omicron variant, which currently dominates the pandemic, prefers the endo/lysosomal cysteine protease cathepsin L over TMPRSS2 for cell entry, raising doubts as to whether TMPRSS2 inhibitors would be suitable for the treatment of patients infected with the Omicron variant. Nevertheless, the contribution of TMPRSS2 to the spread of SARS-CoV-2 in the infected host is largely unclear. In this study, we show that the loss of TMPRSS2 strongly reduced the replication of the Beta variant in the nose, trachea and lung of C57BL/6 mice, and protected the animals from weight loss and disease. The infection of mice with the Omicron variant did not cause disease, as expected, but again, TMPRSS2 was essential for efficient viral spread in the upper and lower respiratory tract. These results identify the key role of TMPRSS2 in SARS-CoV-2 Beta and Omicron infection, and highlight TMPRSS2 as an attractive target for antiviral intervention.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Humans , Mice , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Mice, Inbred C57BL , Pandemics , Serine Endopeptidases/genetics
6.
PLoS Pathog ; 18(1): e1010219, 2022 01.
Article in English | MEDLINE | ID: mdl-35025971

ABSTRACT

Excessive inflammation is a major cause of morbidity and mortality in many viral infections including influenza. Therefore, there is a need for therapeutic interventions that dampen and redirect inflammatory responses and, ideally, exert antiviral effects. Itaconate is an immunomodulatory metabolite which also reprograms cell metabolism and inflammatory responses when applied exogenously. We evaluated effects of endogenous itaconate and exogenous application of itaconate and its variants dimethyl- and 4-octyl-itaconate (DI, 4OI) on host responses to influenza A virus (IAV). Infection induced expression of ACOD1, the enzyme catalyzing itaconate synthesis, in monocytes and macrophages, which correlated with viral replication and was abrogated by DI and 4OI treatment. In IAV-infected mice, pulmonary inflammation and weight loss were greater in Acod1-/- than in wild-type mice, and DI treatment reduced pulmonary inflammation and mortality. The compounds reversed infection-triggered interferon responses and modulated inflammation in human cells supporting non-productive and productive infection, in peripheral blood mononuclear cells, and in human lung tissue. All three itaconates reduced ROS levels and STAT1 phosphorylation, whereas AKT phosphorylation was reduced by 4OI and DI but increased by itaconate. Single-cell RNA sequencing identified monocytes as the main target of infection and the exclusive source of ACOD1 mRNA in peripheral blood. DI treatment silenced IFN-responses predominantly in monocytes, but also in lymphocytes and natural killer cells. Ectopic synthesis of itaconate in A549 cells, which do not physiologically express ACOD1, reduced infection-driven inflammation, and DI reduced IAV- and IFNγ-induced CXCL10 expression in murine macrophages independent of the presence of endogenous ACOD1. The compounds differed greatly in their effects on cellular gene homeostasis and released cytokines/chemokines, but all three markedly reduced release of the pro-inflammatory chemokines CXCL10 (IP-10) and CCL2 (MCP-1). Viral replication did not increase under treatment despite the dramatically repressed IFN responses. In fact, 4OI strongly inhibited viral transcription in peripheral blood mononuclear cells, and the compounds reduced viral titers (4OI>Ita>DI) in A549 cells whereas viral transcription was unaffected. Taken together, these results reveal itaconates as immunomodulatory and antiviral interventions for influenza virus infection.


Subject(s)
Influenza A virus/immunology , Macrophages/immunology , Orthomyxoviridae Infections/drug therapy , Succinates/pharmacology , A549 Cells , Animals , Carboxy-Lyases/deficiency , Carboxy-Lyases/immunology , Cytokines/genetics , Cytokines/immunology , Humans , Macrophages/virology , Mice , Mice, Knockout , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , THP-1 Cells
7.
Cell Host Microbe ; 29(11): 1663-1679.e7, 2021 11 10.
Article in English | MEDLINE | ID: mdl-34610293

ABSTRACT

Gut colonization with multidrug-resistant (MDR) bacteria enhances the risk of bloodstream infections in susceptible individuals. We demonstrate highly variable degrees of ex vivo colonization resistance against a carbapenem-resistant Klebsiella pneumoniae strain in human feces samples and subsequently isolate diverse K. oxytoca strains from protected donors. Several of these K. oxytoca strains reduce gut colonization of MDR K. pneumoniae strains in antibiotic-treated and gnotobiotic mouse models. Comparative analysis of K. oxytoca strains coupled with CRISPR-Cas9-mediated deletion of casA, a protein essential for utilization of selected beta-glucosides, identified competition for specific carbohydrates as key in promoting colonization resistance. In addition to direct competition between K. oxytoca and K. pneumoniae, cooperation with additional commensals is required to reestablish full colonization resistance and gut decolonization. Finally, humanized microbiota mice generated from K. pneumoniae-susceptible donors are protected by K. oxytoca administration, demonstrating the potential of commensal K. oxytoca strains as next-generation probiotics.


Subject(s)
Carbohydrate Metabolism , Feces/microbiology , Gastrointestinal Tract/microbiology , Klebsiella oxytoca/physiology , Klebsiella pneumoniae/growth & development , Microbial Interactions , Adaptive Immunity , Adult , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Child , Drug Resistance, Multiple, Bacterial , Gastrointestinal Microbiome , Germ-Free Life , Glucosides/metabolism , Humans , Klebsiella Infections/immunology , Klebsiella Infections/microbiology , Klebsiella oxytoca/genetics , Klebsiella oxytoca/isolation & purification , Klebsiella pneumoniae/drug effects , Mice , Mice, Inbred C57BL
8.
Front Immunol ; 12: 705436, 2021.
Article in English | MEDLINE | ID: mdl-34512629

ABSTRACT

Autophagy is an evolutionary conserved catabolic pathway that ensures the degradation of intracellular components. The autophagic pathway is regulated by autophagy-related (Atg) proteins that govern formation of double-membraned vesicles called autophagosomes. Autophagy deficiency in regulatory T (Treg) cells leads to increased apoptosis of these cells and to the development of autoimmune disorders, predominantly characterized by intestinal inflammation. Recently, RORγt-expressing Treg cells have been identified as key regulators of gut homeostasis, preventing intestinal immunopathology. To study the role of autophagy in RORγt+ Foxp3+ Treg cells, we generated mice lacking the essential component of the core autophagy machinery Atg5 in Foxp3+ cells. Atg5 deficiency in Treg cells led to a predominant intestinal inflammation. While Atg5-deficient Treg cells were reduced in peripheral lymphoid organs, the intestinal RORγt+ Foxp3+ subpopulation of Treg cells was most severely affected. Our data indicated that autophagy is essential to maintain the intestinal RORγt+ Foxp3+ Treg population, thereby protecting the mice from gut inflammatory disorders.


Subject(s)
Autophagy-Related Protein 5/immunology , Gastrointestinal Microbiome/immunology , Homeostasis/immunology , Intestinal Mucosa/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autophagy-Related Protein 5/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Homeostasis/genetics , Mice , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3
9.
Commun Biol ; 4(1): 47, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420317

ABSTRACT

The occurrence and spread of multidrug-resistant pathogens, especially bacteria from the ESKAPE panel, increases the risk to succumb to untreatable infections. We developed a novel antimicrobial peptide, Pam-3, with antibacterial and antibiofilm properties to counter this threat. The peptide is based on an eight-amino acid carboxyl-terminal fragment of human ß-defensin 1. Pam-3 exhibited prominent antimicrobial activity against multidrug-resistant ESKAPE pathogens and additionally eradicated already established biofilms in vitro, primarily by disrupting membrane integrity of its target cell. Importantly, prolonged exposure did not result in drug-resistance to Pam-3. In mouse models, Pam-3 selectively reduced acute intestinal Salmonella and established Citrobacter infections, without compromising the core microbiota, hence displaying an added benefit to traditional broad-spectrum antibiotics. In conclusion, our data support the development of defensin-derived antimicrobial agents as a novel approach to fight multidrug-resistant bacteria, where Pam-3 appears as a particularly promising microbiota-preserving candidate.


Subject(s)
Enterobacteriaceae Infections/drug therapy , Gastrointestinal Diseases/drug therapy , Gastrointestinal Microbiome/drug effects , Salmonella Infections, Animal/drug therapy , Animals , Biofilms/drug effects , Disease Models, Animal , Drug Resistance, Multiple, Bacterial , Female , Male , Mice, Inbred C57BL , Microbial Sensitivity Tests
10.
Mucosal Immunol ; 14(1): 113-124, 2021 01.
Article in English | MEDLINE | ID: mdl-32433514

ABSTRACT

Diverse microbial signatures within the intestinal microbiota have been associated with intestinal and systemic inflammatory diseases, but whether these candidate microbes actively modulate host phenotypes or passively expand within the altered microbial ecosystem is frequently not known. Here we demonstrate that colonization of mice with a member of the genus Prevotella, which has been previously associated to colitis in mice, exacerbates intestinal inflammation. Our analysis revealed that Prevotella intestinalis alters composition and function of the ecosystem resulting in a reduction of short-chain fatty acids, specifically acetate, and consequently a decrease in intestinal IL-18 levels during steady state. Supplementation of IL-18 to Prevotella-colonized mice was sufficient to reduce intestinal inflammation. Hence, we conclude that intestinal Prevotella colonization results in metabolic changes in the microbiota, which reduce IL-18 production and consequently exacerbate intestinal inflammation, and potential systemic autoimmunity.


Subject(s)
Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/microbiology , Gastrointestinal Microbiome/immunology , Host-Pathogen Interactions/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Prevotella/immunology , Adaptive Immunity , Animals , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Metagenome , Metagenomics/methods , Mice , Mice, Knockout , Mucositis/etiology , Mucositis/metabolism , Mucositis/pathology
11.
PLoS Pathog ; 16(3): e1008448, 2020 03.
Article in English | MEDLINE | ID: mdl-32208465

ABSTRACT

The composition of the intestinal microbiota influences the outcome of enteric infections in human and mice. However, the role of specific members and their metabolites contributing to disease severity is largely unknown. Using isogenic mouse lines harboring distinct microbiota communities, we observed highly variable disease kinetics of enteric Citrobacter rodentium colonization after infection. Transfer of communities from susceptible and resistant mice into germ-free mice verified that the varying susceptibilities are determined by microbiota composition. The strongest differences in colonization were observed in the cecum and could be maintained in vitro by coculturing cecal bacteria with C. rodentium. Cohousing of animals as well as the transfer of cultivable bacteria from resistant to susceptible mice led to variable outcomes in the recipient mice. Microbiome analysis revealed that a higher abundance of butyrate-producing bacteria was associated with the resistant phenotype. Quantification of short-chain fatty acid (SCFA) levels before and after infection revealed increased concentrations of acetate, butyrate and propionate in mice with delayed colonization. Addition of physiological concentrations of butyrate, but not of acetate and/or propionate strongly impaired growth of C. rodentium in vitro. In vivo supplementation of susceptible, antibiotic-treated and germ-free mice with butyrate led to the same level of protection, notably only when cecal butyrate concentration reached a concentration higher than 50 nmol/mg indicating a critical threshold for protection. In the recent years, commensal-derived primary and secondary bacterial metabolites emerged as potent modulators of hosts susceptibility to infection. Our results provide evidence that variations in SCFA production in mice fed fibre-rich chow-based diets modulate susceptibility to colonization with Enterobacteriaceae not only in antibiotic-disturbed ecosystems but even in undisturbed microbial communities. These findings emphasise the need for microbiota normalization across laboratory mouse lines for infection experiments with the model-pathogen C. rodentium independent of investigations of diet and antibiotic usage.


Subject(s)
Citrobacter rodentium/growth & development , Enterobacteriaceae Infections/metabolism , Fatty Acids/metabolism , Gastrointestinal Microbiome , Animals , Mice
12.
Article in English | MEDLINE | ID: mdl-32015030

ABSTRACT

Infections with enterohemorrhagic Escherichia coli (EHEC) cause disease ranging from mild diarrhea to hemolytic-uremic syndrome (HUS) and are the most common cause of renal failure in children in high-income countries. The severity of the disease derives from the release of Shiga toxins (Stx). The use of antibiotics to treat EHEC infections is generally avoided, as it can result in increased stx expression. Here, we systematically tested different classes of antibiotics and found that their influence on stx expression and release varies significantly. We assessed a selection of these antibiotics in vivo using the Citrobacter rodentium ϕstx2dact mouse model and show that stx2d-inducing antibiotics resulted in weight loss and kidney damage despite clearance of the infection. However, several non-Stx-inducing antibiotics cleared bacterial infection without causing Stx-mediated pathology. Our results suggest that these antibiotics might be useful in the treatment of EHEC-infected human patients and decrease the risk of HUS development.


Subject(s)
Acute Kidney Injury/prevention & control , Anti-Bacterial Agents/therapeutic use , Enterohemorrhagic Escherichia coli/drug effects , Escherichia coli Infections/drug therapy , Shiga Toxin 2/metabolism , Acute Kidney Injury/microbiology , Animals , Citrobacter rodentium/genetics , Citrobacter rodentium/metabolism , Disease Models, Animal , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Female , Hemolytic-Uremic Syndrome/drug therapy , Hemolytic-Uremic Syndrome/microbiology , Mice , Mice, Inbred C57BL , Shiga Toxin 2/genetics , Shiga Toxin 2/toxicity
13.
Front Immunol ; 11: 592328, 2020.
Article in English | MEDLINE | ID: mdl-33613516

ABSTRACT

Chronic hepatotropic viral infections are characterized by exhausted CD8+ T cells in the presence of cognate antigen in the liver. The impairment of T cell response limits the control of chronic hepatotropic viruses. Immune-modulatory strategies are attractive options to re-invigorate exhausted T cells. However, in hepatotropic viral infections, the knowledge about immune-modulatory effects on the in-situ regulation of exhausted intrahepatic CD8+ T cells is limited. In this study, we elucidated the functional heterogeneity in the pool of exhausted CD8+ T cells in the liver of mice expressing the model antigen Ova in a fraction of hepatocytes. We found a subpopulation of intrahepatic CXCR5+ Ova-specific CD8+ T cells, which are profoundly cytotoxic, exhibiting efficient metabolic functions as well as improved memory recall and self-maintenance. The intrahepatic Ova-specific CXCR5+ CD8+ T cells are possibly tissue resident cells, which may rely largely on OXPHOS and glycolysis to fuel their cellular processes. Importantly, host conditioning with CpG oligonucleotide reinvigorates and promotes exhausted T cell expansion, facilitating complete antigen eradication. The CpG oligonucleotide-mediated reinvigoration may support resident memory T cell formation and the maintenance of CXCR5+ Ova-specific CD8+ T cells in the liver. These findings suggest that CpG oligodinucleotide may preferentially target CXCR5+ CD8+ T cells for expansion to facilitate the revival of exhausted T cells. Thus, therapeutic strategies aiming to expand CXCR5+ CD8+ T cells might provide a novel approach against chronic liver infection.


Subject(s)
Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunomodulation , Liver/immunology , Liver/metabolism , Receptors, CXCR5/metabolism , Adoptive Transfer , Animals , Biomarkers , Cell Proliferation , Immunization , Liver/pathology , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Mitochondria/immunology , Mitochondria/metabolism , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
14.
PLoS Pathog ; 15(9): e1008036, 2019 09.
Article in English | MEDLINE | ID: mdl-31525249

ABSTRACT

Cytomegalovirus (CMV) is a ubiquitous ß-herpesvirus that establishes life-long latent infection in a high percentage of the population worldwide. CMV induces the strongest and most durable CD8+ T cell response known in human clinical medicine. Due to its unique properties, the virus represents a promising candidate vaccine vector for the induction of persistent cellular immunity. To take advantage of this, we constructed a recombinant murine CMV (MCMV) expressing an MHC-I restricted epitope from influenza A virus (IAV) H1N1 within the immediate early 2 (ie2) gene. Only mice that were immunized intranasally (i.n.) were capable of controlling IAV infection, despite the greater potency of the intraperitoneally (i.p.) vaccination in inducing a systemic IAV-specific CD8+ T cell response. The protective capacity of the i.n. immunization was associated with its ability to induce IAV-specific tissue-resident memory CD8+ T (CD8TRM) cells in the lungs. Our data demonstrate that the protective effect exerted by the i.n. immunization was critically mediated by antigen-specific CD8+ T cells. CD8TRM cells promoted the induction of IFNγ and chemokines that facilitate the recruitment of antigen-specific CD8+ T cells to the lungs. Overall, our results showed that locally applied MCMV vectors could induce mucosal immunity at sites of entry, providing superior immune protection against respiratory infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity, Mucosal , Influenza Vaccines/immunology , Muromegalovirus/immunology , Administration, Intranasal , Amino Acid Sequence , Animals , Cell Line , Chemokines/biosynthesis , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Gene Products, env/administration & dosage , Gene Products, env/genetics , Gene Products, env/immunology , Genetic Vectors , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza, Human/immunology , Influenza, Human/prevention & control , Lung/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Muromegalovirus/genetics , NIH 3T3 Cells , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/virology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
15.
J Am Assoc Lab Anim Sci ; 58(1): 87-91, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30401066

ABSTRACT

The sterilization of potentially infectious animal carcasses is an important biologic safety issue in animal facilities operating as infection or quarantine barriers. However, the literature lacks a validated protocol. Here we describe the validation of an autoclave program suitable for daily use in a small rodent biocontainment unit. We evaluated several procedures for processing mouse carcasses in a standard autoclave. Heat sensors and biologic indicators were implanted inside the peritoneal cavity of dead mice, which were loaded at various densities into IVC cages or metal boxes. Heat sensors revealed broad differences in temperature inside carcasses compared with the autoclave chamber. Achieving the appropriate sterilization temperature was considerably prolonged in carcasses compared with typical laboratory waste material. We show that for 5 cadavers placed well separated inside an IVC, a modified program for mouse cage sterilization using 134 °C for 15 min is suitable. To sterilize approximately 1 kg of carcasses in autoclavable boxes, a period of 6 h is required to reach an effective temperature of 121 °C for 60 min at the center of the waste by using an autoclave program for liquids. In conclusion, we here validated 2 protocols for the sterilization of potentially infectious mouse carcasses, to ensure the application of efficacious procedures.


Subject(s)
Cadaver , Sterilization/methods , Animals , Laboratory Animal Science , Mice , Reproducibility of Results , Temperature
16.
Environ Microbiol ; 21(3): 883-897, 2019 03.
Article in English | MEDLINE | ID: mdl-30411474

ABSTRACT

Pseudomonas aeruginosa is an environmental microorganism and a causative agent of diverse acute and chronic, biofilm-associated infections. Advancing research-based knowledge on its adaptation to conditions within the human host is bound to reveal novel strategies and targets for therapeutic intervention. Here, we investigated the traits that P. aeruginosa PA14 as well as a virulence attenuated ΔlasR mutant need to survive in selected murine infection models. Experimentally, the genetic programs that the bacteria use to adapt to biofilm-associated versus acute infections were dissected by passaging transposon mutant libraries through mouse lungs (acute) or mouse tumours (biofilm-infection). Adaptive metabolic changes of P. aeruginosa were generally required during both infection processes. Counter-selection against flagella expression was observed during acute lung infections. Obviously, avoidance of flagella-mediated activation of host immunity is advantageous for the wildtype bacteria. For the ΔlasR mutant, loss of flagella did not confer a selective advantage. Apparently, other pathogenesis mechanisms are active in this virulence attenuated strain. In contrast, the infective process of P. aeruginosa in the chronic biofilm model apparently required expression of flagellin. Together, our findings imply that the host immune reactions against the infectious agent are very decisive for acuteness and duration of the infectious disease. They direct disease outcome.


Subject(s)
Flagella/physiology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/physiology , Animals , Biofilms , Chronic Disease , Flagella/genetics , Mice , Mice, Inbred BALB C , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Respiratory Tract Infections/microbiology , Virulence
17.
Front Immunol ; 9: 1424, 2018.
Article in English | MEDLINE | ID: mdl-29988532

ABSTRACT

Streptococcus pneumoniae is the most common cause of community-acquired pneumonia (CAP). Despite the low prevalence of CAP caused by methicillin-resistant Staphylococcus aureus (MRSA), CAP patients often receive empirical antibiotic therapy providing coverage for MRSA such as vancomycin or linezolid. An early differentiation between S. pneumoniae and S. aureus pneumonia can help to reduce the use of unnecessary antibiotics. The objective of this study was to identify candidate biomarkers that can discriminate pneumococcal from staphylococcal pneumonia. A genome-wide transcriptional analysis of lung and peripheral blood performed in murine models of S. pneumoniae and S. aureus lung infection identified an interferon signature specifically associated with S. pneumoniae infection. Prediction models built using a support vector machine and Monte Carlo cross-validation, identified the combination of the interferon-induced chemokines CXCL9 and CXCL10 serum concentrations as the set of biomarkers with best sensitivity, specificity, and predictive power that enabled an accurate discrimination between S. pneumoniae and S. aureus pneumonia. The predictive performance of these biomarkers was further validated in an independent cohort of mice. This study highlights the potential of serum CXCL9 and CXCL10 biomarkers as an adjunctive diagnostic tool that could facilitate prompt and correct pathogen-targeted therapy in CAP patients.

18.
Article in English | MEDLINE | ID: mdl-29707522

ABSTRACT

Background: Group A streptococci may induce lymphopenia, but the value of lymphocyte loss as early biomarkers for systemic spread and severe infection has not been examined systematically. Methods: We evaluated peripheral blood cell indices as biomarkers for severity and spread of infection in a mouse model of Streptococcus pyogenes skin infection, using two isolates of greatly differing virulence. Internal organs were examined histologically. Results: After subcutaneous inoculation, strain AP1 disseminated rapidly to peripheral blood and internal organs, causing frank sepsis. In contrast, seeding of internal organs by 5448 was mild, this strain could not be isolated from blood, and infection remained mostly localized to skin. Histopathologic examination of liver revealed microvesicular fatty change (steatosis) in AP1 infection, and examination of spleen showed elevated apoptosis and blurring of the white pulp/red pulp border late (40 h post infection) in AP1 infection. Both strains caused profound lymphopenia, but lymphocyte loss was more rapid early in AP1 infection, and lymphocyte count at 6 h post infection was the most accurate early marker for AP1 infection (area under the receiver operator curve [AUC] = 0.93), followed by the granulocyte/lymphocyte ratio (AUC = 0.89). Conclusions: The results suggest that virulence of S. pyogenes correlates with the degree of early lymphopenia and underscore the value of peripheral blood indices to predict severity of bacterial infections in mice. Early lymphopenia and elevated granulocyte/lymphocyte ratio merit further investigation as biomarkers for systemic spread of S. pyogenes skin infections in humans and, possibly, related pyogenic streptococci in humans and animals.


Subject(s)
Bacterial Load , Granulocytes/cytology , Lymphocytes/cytology , Lymphopenia/microbiology , Streptococcal Infections/immunology , Streptococcus pyogenes/pathogenicity , Animals , Biomarkers/blood , Disease Models, Animal , Mice , Mice, Inbred C57BL , Sepsis/microbiology , Skin/microbiology , Skin/pathology , Streptococcal Infections/microbiology , Streptococcus pyogenes/immunology , Virulence
19.
Cell Rep ; 21(4): 994-1008, 2017 Oct 24.
Article in English | MEDLINE | ID: mdl-29069606

ABSTRACT

Inflammatory bowel disease comprises a group of heterogeneous diseases characterized by chronic and relapsing mucosal inflammation. Alterations in microbiota composition have been proposed to contribute to disease development, but no uniform signatures have yet been identified. Here, we compare the ability of a diverse set of microbial communities to exacerbate intestinal inflammation after chemical damage to the intestinal barrier. Strikingly, genetically identical wild-type mice differing only in their microbiota composition varied strongly in their colitis susceptibility. Transfer of distinct colitogenic communities in gene-deficient mice revealed that they triggered disease via opposing pathways either independent or dependent on adaptive immunity, specifically requiring antigen-specific CD4+ T cells. Our data provide evidence for the concept that microbial communities may alter disease susceptibility via different immune pathways despite eventually resulting in similar host pathology. This suggests a potential benefit for personalizing IBD therapies according to patient-specific microbiota signatures.


Subject(s)
Adaptive Immunity , Colitis, Ulcerative/microbiology , Gastrointestinal Microbiome , Immunity, Innate , Animals , CD4-Positive T-Lymphocytes/immunology , Colitis, Ulcerative/immunology , Female , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL
20.
Eur J Microbiol Immunol (Bp) ; 7(2): 112-126, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28690878

ABSTRACT

Using quantitative phosphopeptide sequencing of unstimulated versus stimulated primary murine Foxp3+ regulatory and Foxp3- conventional T cells (Tregs and Tconv, respectively), we detected a novel and differentially regulated tyrosine phosphorylation site within the C1 domain of the guanine-nucleotide exchange factor CalDAG GEFI. We hypothesized that the Treg-specific and activation-dependent reduced phosphorylation at Y523 allows binding of CalDAG GEFI to diacylglycerol, thereby impacting the formation of a Treg-specific immunological synapse. However, diacylglycerol binding assays of phosphomutant C1 domains of CalDAG GEFI could not confirm this hypothesis. Moreover, CalDAG GEFI-/- mice displayed normal Treg numbers in thymus and secondary lymphoid organs, and CalDAG GEFI-/- Tregs showed unaltered in vitro suppressive capacity when compared to CalDAG GEFI+/+ Tregs. Interestingly, when tested in vivo, CalDAG GEFI-/- Tregs displayed a slightly reduced suppressive ability in the transfer colitis model when compared to CalDAG GEFI+/+ Tregs. Additionally, CRISPR-Cas9-generated CalDAG GEFI-/- Jurkat T cell clones showed reduced adhesion to ICAM-1 and fibronectin when compared to CalDAG GEFI-competent Jurkat T cells. Therefore, we speculate that deficiency in CalDAG GEFI impairs adherence of Tregs to antigen-presenting cells, thereby impeding formation of a fully functional immunological synapse, which finally results in a reduced suppressive potential.

SELECTION OF CITATIONS
SEARCH DETAIL