Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 246, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38172096

ABSTRACT

Members of the low-density lipoprotein receptor (LDLR) family, including LDLRAD3, VLDLR, and ApoER2, were recently described as entry factors for different alphaviruses. However, based on studies with gene edited cells and knockout mice, blockade or abrogation of these receptors does not fully inhibit alphavirus infection, indicating the existence of additional uncharacterized entry factors. Here, we perform a CRISPR-Cas9 genome-wide loss-of-function screen in mouse neuronal cells with a chimeric alphavirus expressing the Eastern equine encephalitis virus (EEEV) structural proteins and identify LDLR as a candidate receptor. Expression of LDLR on the surface of neuronal or non-neuronal cells facilitates binding and infection of EEEV, Western equine encephalitis virus, and Semliki Forest virus. Domain mapping and binding studies reveal a low-affinity interaction with LA domain 3 (LA3) that can be enhanced by concatenation of LA3 repeats. Soluble decoy proteins with multiple LA3 repeats inhibit EEEV infection in cell culture and in mice. Our results establish LDLR as a low-affinity receptor for multiple alphaviruses and highlight a possible path for developing inhibitors that could mitigate infection and disease.


Subject(s)
Alphavirus Infections , Alphavirus , Encephalitis Virus, Eastern Equine , Horses , Animals , Mice , Alphavirus/genetics , Encephalitis Virus, Eastern Equine/genetics , Semliki forest virus/genetics , Lipoproteins, LDL
2.
Nat Commun ; 14(1): 5973, 2023 09 25.
Article in English | MEDLINE | ID: mdl-37749080

ABSTRACT

The determinants of severe disease caused by West Nile virus (WNV) and why only ~1% of individuals progress to encephalitis remain poorly understood. Here, we use human and mouse enteroids, and a mouse model of pathogenesis, to explore the capacity of WNV to directly infect gastrointestinal (GI) tract cells and contribute to disease severity. At baseline, WNV poorly infects human and mouse enteroid cultures and enterocytes in mice. However, when STAT1 or type I interferon (IFN) responses are absent, GI tract cells become infected, and this is associated with augmented GI tract and blood-brain barrier (BBB) permeability, accumulation of gut-derived molecules in the brain, and more severe WNV disease. The increased gut permeability requires TNF-α signaling, and is absent in WNV-infected IFN-deficient germ-free mice. To link these findings to human disease, we measured auto-antibodies against type I IFNs in serum from WNV-infected human cohorts. A greater frequency of auto- and neutralizing antibodies against IFN-α2 or IFN-ω is present in patients with severe WNV infection, whereas virtually no asymptomatic WNV-infected subjects have such antibodies (odds ratio 24 [95% confidence interval: 3.0 - 192.5; P = 0.003]). Overall, our experiments establish that blockade of type I IFN signaling extends WNV tropism to enterocytes, which correlates with increased gut and BBB permeability, and more severe disease.


Subject(s)
Interferon Type I , West Nile Fever , West Nile virus , Humans , Animals , Mice , Brain , Antibodies, Neutralizing
3.
Cell Rep ; 42(8): 112946, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37556325

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is an encephalitic alphavirus responsible for epidemics of neurological disease across the Americas. Low-density lipoprotein receptor class A domain-containing 3 (LDLRAD3) is a recently reported entry receptor for VEEV. Here, using wild-type and Ldlrad3-deficient mice, we define a critical role for LDLRAD3 in controlling steps in VEEV infection, pathogenesis, and neurotropism. Our analysis shows that LDLRAD3 is required for efficient VEEV infection and pathogenesis prior to and after central nervous system invasion. Ldlrad3-deficient mice survive intranasal and intracranial VEEV inoculation and show reduced infection of neurons in different brain regions. As LDLRAD3 is a determinant of pathogenesis and an entry receptor required for VEEV infection of neurons of the brain, receptor-targeted therapies may hold promise as countermeasures.


Subject(s)
Encephalomyelitis, Venezuelan Equine , Receptors, LDL , Animals , Mice , Brain/pathology , Central Nervous System , Encephalitis Virus, Venezuelan Equine/physiology , Encephalomyelitis, Venezuelan Equine/pathology , Receptors, LDL/physiology
4.
Cell ; 184(5): 1214-1231.e16, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33636133

ABSTRACT

Although enteric helminth infections modulate immunity to mucosal pathogens, their effects on systemic microbes remain less established. Here, we observe increased mortality in mice coinfected with the enteric helminth Heligmosomoides polygyrus bakeri (Hpb) and West Nile virus (WNV). This enhanced susceptibility is associated with altered gut morphology and transit, translocation of commensal bacteria, impaired WNV-specific T cell responses, and increased virus infection in the gastrointestinal tract and central nervous system. These outcomes were due to type 2 immune skewing, because coinfection in Stat6-/- mice rescues mortality, treatment of helminth-free WNV-infected mice with interleukin (IL)-4 mirrors coinfection, and IL-4 receptor signaling in intestinal epithelial cells mediates the susceptibility phenotypes. Moreover, tuft cell-deficient mice show improved outcomes with coinfection, whereas treatment of helminth-free mice with tuft cell-derived cytokine IL-25 or ligand succinate worsens WNV disease. Thus, helminth activation of tuft cell-IL-4-receptor circuits in the gut exacerbates infection and disease of a neurotropic flavivirus.


Subject(s)
Coinfection , Nematospiroides dubius/physiology , Signal Transduction , Strongylida Infections/pathology , West Nile virus/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Disease Susceptibility , Intestinal Mucosa/parasitology , Intestinal Mucosa/virology , Mice , Mice, Inbred C57BL , Neurons/parasitology , Neurons/virology , Receptors, Interleukin-4/metabolism , STAT6 Transcription Factor/genetics , Severity of Illness Index , Strongylida Infections/parasitology
5.
Front Mol Neurosci ; 13: 149, 2020.
Article in English | MEDLINE | ID: mdl-33132837

ABSTRACT

A growing body of evidence indicates that microglia actively remove synapses in vivo, thereby playing a key role in synaptic refinement and modulation of brain connectivity. This phenomenon was mainly investigated in immunofluorescence staining and confocal microscopy. However, a quantification of synaptic material in microglia using these techniques is extremely time-consuming and labor-intensive. To address this issue, we aimed to quantify synaptic proteins in microglia using flow cytometry. With this approach, we first showed that microglia from the healthy adult mouse brain contain a detectable level of VGLUT1 protein. Next, we found more than two-fold increased VGLUT1 immunoreactivity in microglia from the developing brain (P15) as compared to adult microglia. These data indicate that microglia-mediated synaptic pruning mostly occurs during the brain developmental period. We then quantified the VGLUT1 staining in microglia in two transgenic models characterized by pathological microglia-mediated synaptic pruning. In the 5xFAD mouse model of Alzheimer's disease (AD) microglia exhibited a significant increase in VGLUT1 immunoreactivity before the onset of amyloid pathology. Moreover, conditional deletion of TDP-43 in microglia, which causes a hyper-phagocytic phenotype associated with synaptic loss, also resulted in increased VGLUT1 immunoreactivity within microglia. This work provides a quantitative assessment of synaptic proteins in microglia, under homeostasis, and in mouse models of disease.

6.
Cell ; 179(5): 1144-1159.e15, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31708126

ABSTRACT

The colonic epithelium can undergo multiple rounds of damage and repair, often in response to excessive inflammation. The responsive stem cell that mediates this process is unclear, in part because of a lack of in vitro models that recapitulate key epithelial changes that occur in vivo during damage and repair. Here, we identify a Hopx+ colitis-associated regenerative stem cell (CARSC) population that functionally contributes to mucosal repair in mouse models of colitis. Hopx+ CARSCs, enriched for fetal-like markers, transiently arose from hypertrophic crypts known to facilitate regeneration. Importantly, we established a long-term, self-organizing two-dimensional (2D) epithelial monolayer system to model the regenerative properties and responses of Hopx+ CARSCs. This system can reenact the "homeostasis-injury-regeneration" cycles of epithelial alterations that occur in vivo. Using this system, we found that hypoxia and endoplasmic reticulum stress, insults commonly present in inflammatory bowel diseases, mediated the cyclic switch of cellular status in this process.


Subject(s)
Cell Culture Techniques/methods , Colon/pathology , Stem Cells/pathology , 3T3 Cells , Animals , Colitis/pathology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Homeodomain Proteins/metabolism , Mice , Models, Biological , Oxygen/pharmacology , Regeneration/drug effects , Stem Cells/drug effects , Stress, Physiological/drug effects
7.
FASEB J ; 33(7): 8634-8647, 2019 07.
Article in English | MEDLINE | ID: mdl-31090455

ABSTRACT

Reduced expression of 2'-3'-cyclic nucleotide 3'-phosphodiesterase (Cnp) in humans and mice causes white matter inflammation and catatonic signs. These consequences are experimentally alleviated by microglia ablation via colony-stimulating factor 1 receptor (CSF1R) inhibition using PLX5622. Here we address for the first time preclinical topics crucial for translation, most importantly 1) the comparison of 2 long-term PLX5622 applications (prevention and treatment) vs. 1 treatment alone, 2) the correlation of catatonic signs and executive dysfunction, 3) the phenotype of leftover microglia evading depletion, and 4) the role of intercellular interactions for efficient CSF1R inhibition. Based on our Cnp-/- mouse model and in vitro time-lapse imaging, we report the unexpected discovery that microglia surviving under PLX5622 display a highly inflammatory phenotype including aggressive premortal phagocytosis of oligodendrocyte precursor cells. Interestingly, ablating microglia in vitro requires mixed glial cultures, whereas cultured pure microglia withstand PLX5622 application. Importantly, 2 extended rounds of CSF1R inhibition are not superior to 1 treatment regarding any readout investigated (magnetic resonance imaging and magnetic resonance spectroscopy, behavior, immunohistochemistry). Catatonia-related executive dysfunction and brain atrophy of Cnp-/- mice fail to improve under PLX5622. To conclude, even though microglia depletion is temporarily beneficial and worth pursuing, complementary treatment strategies are needed for full and lasting recovery.-Fernandez Garcia-Agudo, L., Janova, H., Sendler, L. E., Arinrad, S., Steixner, A. A., Hassouna, I., Balmuth, E., Ronnenberg, A., Schopf, N., van der Flier, F. J., Begemann, M., Martens, H., Weber, M. S., Boretius, S., Nave, K.-A., Ehrenreich, H. Genetically induced brain inflammation by Cnp deletion transiently benefits from microglia depletion.


Subject(s)
2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/genetics , Brain/pathology , Encephalitis/genetics , Microglia/pathology , Sequence Deletion/genetics , Adult , Animals , Brain/drug effects , Female , Humans , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Organic Chemicals/pharmacology , Phenotype , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Sequence Deletion/drug effects
8.
Glia ; 66(5): 920-933, 2018 05.
Article in English | MEDLINE | ID: mdl-29350438

ABSTRACT

Human mesial temporal lobe epilepsy (MTLE) features subregion-specific hippocampal neurodegeneration and reactive astrogliosis, including up-regulation of the glial fibrillary acidic protein (GFAP) and down-regulation of glutamine synthetase (GS). However, the regional astrocytic expression pattern of GFAP and GS upon MTLE-associated neurodegeneration still remains elusive. We assessed GFAP and GS expression in strict correlation with the local neuronal number in cortical and hippocampal surgical specimens from 16 MTLE patients using immunohistochemistry, stereology and high-resolution image analysis for digital pathology and whole-slide imaging. In the cortex, GS-positive (GS+) astrocytes are dominant in all neuronal layers, with a neuron to GS+ cell ratio of 2:1. GFAP-positive (GFAP+) cells are widely spaced, with a GS+ to GFAP+ cell ratio of 3:1-5:1. White matter astrocytes, on the contrary, express mainly GFAP and, to a lesser extent, GS. In the hippocampus, the neuron to GS+ cell ratio is approximately 1:1. Hippocampal degeneration is associated with a reduction of GS+ astrocytes, which is proportional to the degree of neuronal loss and primarily present in the hilus. Up-regulation of GFAP as a classical hallmark of reactive astrogliosis does not follow the GS-pattern and is prominent in the CA1. Reactive alterations were proportional to the neuronal loss in the neuronal somatic layers (stratum pyramidale and hilus), while observed to a lesser extent in the axonal/dendritic layers (stratum radiatum, molecular layer). We conclude that astrocytic GS is expressed in the neuronal somatic layers and, upon neurodegeneration, is down-regulated proportionally to the degree of neuronal loss.


Subject(s)
Astrocytes/enzymology , Cerebral Cortex/enzymology , Epilepsy, Temporal Lobe/enzymology , Glutamate-Ammonia Ligase/metabolism , Neurons/enzymology , Adult , Astrocytes/pathology , Cell Death/physiology , Cerebral Cortex/pathology , Drug Resistant Epilepsy/enzymology , Drug Resistant Epilepsy/pathology , Drug Resistant Epilepsy/surgery , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/surgery , Female , Glial Fibrillary Acidic Protein/metabolism , Gliosis/enzymology , Gliosis/pathology , Humans , Immunohistochemistry , Male , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology , Neurons/pathology , White Matter/enzymology , White Matter/pathology
9.
J Clin Invest ; 128(2): 734-745, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29252214

ABSTRACT

The underlying cellular mechanisms of catatonia, an executive "psychomotor" syndrome that is observed across neuropsychiatric diseases, have remained obscure. In humans and mice, reduced expression of the structural myelin protein CNP is associated with catatonic signs in an age-dependent manner, pointing to the involvement of myelin-producing oligodendrocytes. Here, we showed that the underlying cause of catatonic signs is the low-grade inflammation of white matter tracts, which marks a final common pathway in Cnp-deficient and other mutant mice with minor myelin abnormalities. The inhibitor of CSF1 receptor kinase signaling PLX5622 depleted microglia and alleviated the catatonic symptoms of Cnp mutants. Thus, microglia and low-grade inflammation of myelinated tracts emerged as the trigger of a previously unexplained mental condition. We observed a very high (25%) prevalence of individuals with catatonic signs in a deeply phenotyped schizophrenia sample (n = 1095). Additionally, we found the loss-of-function allele of a myelin-specific gene (CNP rs2070106-AA) associated with catatonia in 2 independent schizophrenia cohorts and also associated with white matter hyperintensities in a general population sample. Since the catatonic syndrome is likely a surrogate marker for other executive function defects, we suggest that microglia-directed therapies may be considered in psychiatric disorders associated with myelin abnormalities.


Subject(s)
2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/genetics , Catatonia/pathology , Microglia/cytology , Myelin Sheath/chemistry , Adult , Age Factors , Alleles , Animals , Brain/pathology , Catatonia/prevention & control , Female , Genotype , Humans , Inflammation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mutation , Oligodendroglia/cytology , Organic Chemicals/chemistry , Phenotype , Prevalence , Receptor, Macrophage Colony-Stimulating Factor/genetics , Schizophrenia/genetics , White Matter/pathology
10.
Glia ; 65(7): 1176-1185, 2017 07.
Article in English | MEDLINE | ID: mdl-28471051

ABSTRACT

Microglia as principle innate immune cells of the central nervous system (CNS) are the first line of defense against invading pathogens. They are capable of sensing infections through diverse receptors, such as Toll-like receptor 4 (TLR4). This receptor is best known for its ability to recognize bacterial lipopolysaccharide (LPS), a causative agent of gram-negative sepsis and septic shock. A putative, naturally occurring antagonist of TLR4 derives from the photosynthetic bacterium Rhodobacter sphaeroides. However, the antagonistic potential of R. sphaeroides LPS (Rs-LPS) is no universal feature, since several studies suggested agonistic rather than antagonistic actions of this molecule depending on the investigated mammalian species. Here we show the agonistic versus antagonistic potential of Rs-LPS in primary mouse microglia. We demonstrate that Rs-LPS efficiently induces the release of cytokines and chemokines, which depends on TLR4, MyD88, and TRIF, but not CD14. Furthermore, Rs-LPS is able to regulate the phagocytic capacity of microglia as agonist, while it antagonizes Re-LPS-induced MHC I expression. Finally, to our knowledge, we are the first to provide in vivo evidence for an agonistic potential of Rs-LPS, as it efficiently triggers the recruitment of peripheral immune cells to the endotoxin-challenged CNS. Together, our results argue for a versatile and complex organization of the microglial TLR4 system, which specifically translates exogenous signals into cellular functions. Importantly, as demonstrated here for microglia, the antagonistic potential of Rs-LPS needs to be considered with caution, as reactions to Rs-LPS not only differ by cell type, but even by function within one cell type.


Subject(s)
Lipopolysaccharides/pharmacology , Microglia/drug effects , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Animals, Newborn , Brain/cytology , Cells, Cultured , Corpus Striatum/drug effects , Cytokines/metabolism , Dose-Response Relationship, Drug , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Macrophages/drug effects , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/drug effects , Myelin Sheath/pathology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Phagocytosis/drug effects , Phagocytosis/physiology , Toll-Like Receptor 4/genetics , Up-Regulation/drug effects , Up-Regulation/physiology
11.
Glia ; 64(4): 635-49, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26683584

ABSTRACT

Microglia, innate immune cells of the CNS, sense infection and damage through overlapping receptor sets. Toll-like receptor (TLR) 4 recognizes bacterial lipopolysaccharide (LPS) and multiple injury-associated factors. We show that its co-receptor CD14 serves three non-redundant functions in microglia. First, it confers an up to 100-fold higher LPS sensitivity compared to peripheral macrophages to enable efficient proinflammatory cytokine induction. Second, CD14 prevents excessive responses to massive LPS challenges via an interferon ß-mediated feedback. Third, CD14 is mandatory for microglial reactions to tissue damage-associated signals. In mice, these functions are essential for balanced CNS responses to bacterial infection, traumatic and ischemic injuries, since CD14 deficiency causes either hypo- or hyperinflammation, insufficient or exaggerated immune cell recruitment or worsened stroke outcomes. While CD14 orchestrates functions of TLR4 and related immune receptors, it is itself regulated by TLR and non-TLR systems to thereby fine-tune microglial damage-sensing capacity upon infectious and non-infectious CNS challenges.


Subject(s)
Brain Injuries/immunology , Brain Ischemia/immunology , Escherichia coli Infections/metabolism , Lipopolysaccharide Receptors/metabolism , Microglia/immunology , Stroke/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Brain/immunology , Brain/pathology , Brain Injuries/complications , Brain Injuries/pathology , Brain Ischemia/pathology , Cells, Cultured , Disease Models, Animal , Escherichia coli , Escherichia coli Infections/complications , Escherichia coli Infections/pathology , Feedback, Physiological/physiology , Infarction, Middle Cerebral Artery , Interferon-beta/metabolism , Lipopolysaccharide Receptors/genetics , Lipopolysaccharides/toxicity , Macrophages/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Neuroimmunomodulation , Stroke/pathology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
12.
Glia ; 63(6): 1083-99, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25731696

ABSTRACT

The putative protein tyrosine kinase (PTK) inhibitor tyrphostin AG126 has proven beneficial in various models of inflammatory disease. Yet molecular targets and cellular mechanisms remained enigmatic. We demonstrate here that AG126 treatment has beneficial effects in experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis. AG126 alleviates the clinical symptoms, diminishes encephalitogenic Th17 differentiation, reduces inflammatory CNS infiltration as well as microglia activation and attenuates myelin damage. We show that AG126 directly inhibits Bruton's tyrosine kinase (BTK), a PTK associated with B cell receptor and Toll-like receptor (TLR) signaling. However, BTK inhibition cannot account for the entire activity spectrum. Effects on TLR-induced proinflammatory cytokine expression in microglia involve AG126 hydrolysis and conversion of its dinitrile side chain to malononitrile (MN). Notably, while liberated MN can subsequently mediate critical AG126 features, full protection in EAE still requires delivery of intact AG126. Its anti-inflammatory potential and especially interference with TLR signaling thus rely on a dual mechanism encompassing BTK and a novel MN-sensitive target. Both principles bear great potential for the therapeutic management of disturbed innate and adaptive immune functions.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Neuroprotective Agents/pharmacology , Tyrphostins/pharmacology , Agammaglobulinaemia Tyrosine Kinase , Animals , Cells, Cultured , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Hydrolysis , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/physiology , Myeloid Differentiation Factor 88/metabolism , Neuroprotective Agents/chemistry , Nitriles/chemistry , Nitriles/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Spleen/cytology , Spleen/drug effects , Spleen/physiopathology , Th17 Cells/drug effects , Th17 Cells/pathology , Th17 Cells/physiology , Tyrphostins/chemistry
13.
PLoS One ; 9(8): e104064, 2014.
Article in English | MEDLINE | ID: mdl-25084094

ABSTRACT

Adenylate cyclase toxin (CyaA) is a key virulence factor of the whooping cough agent Bordetella pertussis. The toxin targets CD11b-expressing phagocytes and delivers into their cytosol an adenylyl cyclase (AC) enzyme that subverts cellular signaling by increasing cAMP levels. In the present study, we analyzed the modulatory effects of CyaA on adhesive, migratory and antigen presenting properties of Toll-like receptor (TLR)-activated murine and human dendritic cells (DCs). cAMP signaling of CyaA enhanced TLR-induced dissolution of cell adhesive contacts and migration of DCs towards the lymph node-homing chemokines CCL19 and CCL21 in vitro. Moreover, we examined in detail the capacity of toxin-treated DCs to induce CD4(+) and CD8(+) T cell responses. Exposure to CyaA decreased the capacity of LPS-stimulated DCs to present soluble protein antigen to CD4+ T cells independently of modulation of co-stimulatory molecules and cytokine production, and enhanced their capacity to promote CD4(+)CD25(+)Foxp3(+) T regulatory cells in vitro. In addition, CyaA decreased the capacity of LPS-stimulated DCs to induce CD8(+) T cell proliferation and limited the induction of IFN-γ producing CD8(+) T cells while enhancing IL-10 and IL-17-production. These results indicate that through activation of cAMP signaling, the CyaA may be mobilizing DCs impaired in T cell stimulatory capacity and arrival of such DCs into draining lymph nodes may than contribute to delay and subversion of host immune responses during B. pertussis infection.


Subject(s)
Adenylate Cyclase Toxin/pharmacology , Bordetella pertussis/chemistry , Cell Movement/drug effects , Dendritic Cells/cytology , Dendritic Cells/immunology , Lymphocyte Activation/drug effects , Toll-Like Receptors/metabolism , Animals , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion/drug effects , Cell Death/drug effects , Cell Proliferation/drug effects , Dendritic Cells/drug effects , Humans , Mice, Inbred C57BL , Solubility , T-Lymphocytes, Regulatory/drug effects
14.
J Neuroinflammation ; 11: 14, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24456653

ABSTRACT

BACKGROUND: Prophylaxis with unmethylated cytosine phosphate guanidine (CpG) oligodeoxynucleotides (ODN) protects against several systemic experimental infections. Escherichia coli is a major cause of Gram-negative neonatal bacterial meningitis and also causes meningitis and meningoencephalitis in older and immunocompromised patients. METHODS: Wild-type (wt) and Toll-like receptor 9 (TLR9)-deficient mice were rendered neutropenic by intraperitoneal administration of the anti-Ly-6G monoclonal antibody. Immunocompetent and neutropenic mice received intraperitoneal CpG ODN or vehicle 72 h prior to induction of E. coli K1 meningoencephalitis. RESULTS: Pre-treatment with CpG ODN significantly increased survival of neutropenic wt mice from 33% to 75% (P = 0.0003) but did not protect neutropenic TLR9-/- mice. The protective effect of CpG ODN was associated with an enhanced production of interleukin (IL)-12/IL-23p40 with sustained increased levels in serum and spleen at least for 17 days after conditioning compared to buffer-treated animals. CpG-treated neutropenic wt mice showed reduced bacterial concentrations and increased recruitment of Ly6ChighCCR2+ monocytes in brain and spleen 42 h after infection. The levels of macrophage inflammatory protein 1α (MIP-1α) and interferon gamma (IFN-γ) in spleen were higher 42 h after infection in CpG-treated compared to buffer-treated neutropenic animals. In immunocompetent mice, prophylaxis with CpG ODN did not significantly increase survival compared to the buffer group (60% vs. 45%, P = 0.2). CONCLUSIONS: These findings suggest that systemic administration of CpG ODN may help to prevent bacterial CNS infections in immunocompromised individuals.


Subject(s)
Escherichia coli Infections/prevention & control , Guanidine/chemistry , Oligodeoxyribonucleotides/therapeutic use , Animals , Antigens, CD/metabolism , Central Nervous System/drug effects , Central Nervous System/microbiology , Central Nervous System/pathology , Cytokines/metabolism , Disease Models, Animal , Drug Administration Schedule , Escherichia coli/physiology , Flow Cytometry , Meningoencephalitis/prevention & control , Mice , Mice, Knockout , Spleen/microbiology , Spleen/pathology , Toll-Like Receptor 9/deficiency
15.
J Neuroimmunol ; 252(1-2): 16-23, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22889567

ABSTRACT

Increasing the phagocytic activity of microglia could improve the resistance of immunocompromised patients to CNS infections. We studied the microglial responses upon stimulation with the Nod2 ligand muramyl dipeptide (MDP) alone or in combination with a TLR1/2, 3 or 4 agonist. MDP caused a mild release of NO, but induced neither a significant release of pro-inflammatory cytokines nor an expression of molecules associated with professional antigen presentation. Using the Escherichia coli K1 model, microglial pre-stimulation with MDP enhanced bacterial phagocytosis which was strengthened on TLR-pre-stimulated cells. Dual pre-stimulation of Nod2 and TLR1/2 or 4 caused maximal phagocytosis and intracellular killing.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/immunology , Adjuvants, Immunologic , Escherichia coli/immunology , Microglia/immunology , Phagocytosis/immunology , Toll-Like Receptors/immunology , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Adjuvants, Immunologic/pharmacology , Animals , Cells, Cultured , Cytotoxicity, Immunologic/immunology , Escherichia coli Infections/immunology , Flow Cytometry , Immunity, Innate/immunology , Ligands , Mice , Mice, Inbred C57BL , Microglia/drug effects , Nod2 Signaling Adaptor Protein/immunology , Phagocytosis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...