Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 214
Filter
Add more filters

Publication year range
1.
Immunity ; 45(2): 238-9, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27533011

ABSTRACT

Type 3 innate lymphoid cells (ILC3s) and enteric glia, an essential structural component of gut innervation, are well-known regulators of intestinal homeostasis. Ibiza et al. (2016) uncover a new link between commensal bacteria, enteric glial cells, and ILC3s that is required for intestinal homeostasis and defense.


Subject(s)
Dysbiosis/genetics , Gastrointestinal Microbiome/immunology , Immunity, Innate , Intestines/immunology , Lymphocytes/immunology , Neuroglia/immunology , Proto-Oncogene Proteins c-ret/metabolism , Animals , Gene Knockdown Techniques , Glial Cell Line-Derived Neurotrophic Factors/metabolism , Homeostasis , Humans , Interleukins/metabolism , Intestines/innervation , Mice , Neuroglia/microbiology , Proto-Oncogene Proteins c-ret/genetics , Symbiosis , Interleukin-22
2.
J Neuroinflammation ; 16(1): 113, 2019 May 28.
Article in English | MEDLINE | ID: mdl-31138231

ABSTRACT

BACKGROUND: The cellular and molecular pathophysiological mecha\nisms of pain processing in neglected parasitic infections such as leishmaniasis remain unknown. The present study evaluated the participation of spinal cord glial cells in the pathophysiology of pain induced by Leishmania amazonensis infection in BALB/c mice. METHODS: Mice received intra-plantar (i.pl.) injection of L. amazonensis (1 × 105) and hyperalgesia, and paw edema were evaluated bilaterally for 40 days. The levels of TNF-α and IL-1ß, MPO activity, and histopathology were assessed on the 40th day. ATF3 mRNA expression was assessed in DRG cells at the 30th day post-infection. Blood TNF-α and IL-1ß levels and systemic parasite burden were evaluated 5-40 days after the infection. At the 30th day post-infection L. amazonensis, the effects of intrathecal (i.t.) treatments with neutralizing antibody anti-CX3CL1, etanercept (soluble TNFR2 receptor), and interleukin-1 receptor antagonist (IL-1ra) on infection-induced hyperalgesia and paw edema were assessed. In another set of experiments, we performed a time course analysis of spinal cord GFAP and Iba-1 (astrocytes and microglia markers, respectively) and used confocal immunofluorescence and Western blot to confirm the expression at the protein level. Selective astrocyte (α-aminoadipate) and microglia (minocycline) inhibitors were injected i.t. to determine the contribution of these cells to hyperalgesia and paw edema. The effects of i.t. treatments with glial and NFκB (PDTC) inhibitors on spinal glial activation, TNF-α, IL-1ß, CX3CR1 and CX3CL1 mRNA expression, and NFκB activation were also evaluated. Finally, the contribution of TNF-α and IL-1ß to CX3CL1 mRNA expression was investigated. RESULTS: L. amazonensis infection induced chronic mechanical and thermal hyperalgesia and paw edema in the infected paw. Mechanical hyperalgesia was also observed in the contralateral paw. TNF-α, IL-1ß, MPO activity, and epidermal/dermal thickness increased in the infected paw, which confirmed the peripheral inflammation at the primary foci of this infection. ATF3 mRNA expression at the ipsilateral DRG of the infected paw was unaltered 30 days post-infection. TNF-α and IL-1ß blood levels were not changed over the time course of disease, and parasitism increased in a time-dependent manner in the ipsilateral draining lymph node. Treatments targeting CX3CL1, TNF-α, and IL-1ß inhibited L. amazonensis-induced ongoing mechanical and thermal hyperalgesia, but not paw edema. A time course of GFAP, Iba-1, and CX3CR1 mRNA expression indicated spinal activation of astrocytes and microglia, which was confirmed at the GFAP and Iba-1 protein level at the peak of mRNA expression (30th day). Selective astrocyte and microglia inhibition diminished infection-induced ipsilateral mechanical hyperalgesia and thermal hyperalgesia, and contralateral mechanical hyperalgesia, but not ipsilateral paw edema. Targeting astrocytes, microglia and NFκB diminished L. amazonensis-induced GFAP, Iba-1, TNF-α, IL-1ß, CX3CR1 and CX3CL1 mRNA expression, and NFκB activation in the spinal cord at the peak of spinal cord glial cells activation. CX3CL1 mRNA expression was also detected in the ipsilateral DRG of infected mice at the 30th day post-infection, and the i.t. injection of TNF-α or IL-1ß in naïve animals induced CX3CL1 mRNA expression in the spinal cord and ipsilateral DRG. CONCLUSIONS: L. amazonensis skin infection produces chronic pain by central mechanisms involving spinal cord astrocytes and microglia-related production of cytokines and chemokines, and NFκB activation contributes to L. amazonensis infection-induced hyperalgesia and neuroinflammation.


Subject(s)
Edema/pathology , Hyperalgesia/pathology , Leishmaniasis/pathology , Neuroglia/pathology , Pain/pathology , Spinal Cord/pathology , Animals , Edema/microbiology , Hyperalgesia/microbiology , Leishmania , Male , Mice , Mice, Inbred BALB C , Neuroglia/microbiology , Pain/microbiology , Spinal Cord/microbiology
3.
J Biomed Sci ; 26(1): 16, 2019 Feb 07.
Article in English | MEDLINE | ID: mdl-30732602

ABSTRACT

BACKGROUND: Shiga toxin 2 from enterohemorrhagic Escherichia coli is the etiologic agent of bloody diarrhea, hemolytic uremic syndrome and derived encephalopathies that may result to death in patients. Being a Gram negative bacterium, lipopolysaccharide is also released. Particularly, the hippocampus has been found affected in patients intoxicated with Shiga toxin 2. In the current work, the deleterious effects of Shiga toxin 2 and lipopolysaccharide are investigated in detail in hippocampal cells for the first time in a translational murine model, providing conclusive evidences on how these toxins may damage in the observed clinic cases. METHODS: Male NIH mice (25 g) were injected intravenously with saline solution, lipopolysaccharide, Shiga toxin 2 or a combination of Shiga toxin 2 with lipopolysaccharide. Brain water content assay was made to determine brain edema. Another set of animals were intracardially perfused with a fixative solution and their brains were subjected to immunofluorescence with lectins to determine the microvasculature profile, and anti-GFAP, anti-NeuN, anti-MBP and anti-Iba1 to study reactive astrocytes, neuronal damage, myelin dysarrangements and microglial state respectively. Finally, the Thiobarbituric Acid Reactive Substances Assay was made to determine lipid peroxidation. In all assays, statistical significance was performed using the One-way analysis of variance followed by Bonferroni post hoc test. RESULTS: Systemic sublethal administration of Shiga toxin 2 increased the expressions of astrocytic GFAP and microglial Iba1, and decreased the expressions of endothelial glycocalyx, NeuN neurons from CA1 pyramidal layer and oligodendrocytic MBP myelin sheath from the fimbria of the hippocampus. In addition, increased interstitial fluids and Thiobarbituric Acid Reactive Substances-derived lipid peroxidation were also found. The observed outcomes were enhanced when sublethal administration of Shiga toxin 2 was co-administered together with lipopolysaccharide. CONCLUSION: Systemic sublethal administration of Shiga toxin 2 produced a deterioration of the cells that integrate the vascular unit displaying astrocytic and microglial reactive profiles, while edema and lipid peroxidation were also observed. The contribution of lipopolysaccharide to pathogenicity caused by Shiga toxin 2 resulted to enhance the observed hippocampal damage.


Subject(s)
Edema/physiopathology , Enterohemorrhagic Escherichia coli/physiology , Hippocampus/physiopathology , Lipid Peroxidation , Lipopolysaccharides/adverse effects , Shiga Toxin 2/adverse effects , Animals , Edema/microbiology , Hippocampus/drug effects , Hippocampus/microbiology , Lipid Peroxidation/drug effects , Male , Mice , Neuroglia/drug effects , Neuroglia/microbiology , Neuroglia/physiology
4.
Eur J Nutr ; 58(8): 3361-3375, 2019 12.
Article in English | MEDLINE | ID: mdl-30826905

ABSTRACT

PURPOSE: Type 2 diabetes mellitus (T2DM) was associated with gut microbial impairment (dysbiosis) and neurological and behavioral disorders. The role of the gut-brain axis in the management of many diseases including T2DM has been the focus of much research activity in the recent years. However, a wide knowledge gap exists about the gut microbial effects on the function of glia cells. Hence, the present study was aimed to examine the effects of psychobatics on dysbiosis and glia cells function in enteric and central nervous system with an inflammatory insight in T2DM. METHODS: Thirty rats were treated by Lactobacillus (L.) plantarum, inulin, or their combination (synbiotic) for 8 weeks after inducing T2DM. Fecal sample was collected to evaluate gut microbial composition. Then, the rats were sacrificed, and the colon, amygdala, and prefrontal cortex (PFC) were studied. RESULTS: T2DM resulted in dysbiosis and increased levels of glial cell-derived neurotrophic factor (GDNF), glial fibrillary acidic protein (GFAP), and inflammatory markers (IL-17, IL-6, and TLR-2) in the colon and brain. However, concurrent supplementation of L. plantarum and inulin could improve the gut microbial composition as well as reduce the levels of inflammatory cytokines. While the administration of L. plantarum led to a significant decrease in TLR-2 as well as GDNF and GFAP only in the amygdala, the synbiotic intake could make such changes in the colon, amygdala, and PFC. CONCLUSIONS: Our findings demonstrated an innovative approach to the beneficial effects of psychobiotics in neuroinflammation and behavioral performance through gut microbiota changes, focusing on possible role of glial cells in gut-brain axis.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Brain/drug effects , Diabetes Mellitus, Experimental/microbiology , Gastrointestinal Microbiome/drug effects , Inulin/pharmacology , Lactobacillus plantarum , Synbiotics/administration & dosage , Animals , Anti-Anxiety Agents/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Brain/microbiology , Diabetes Mellitus, Experimental/physiopathology , Disease Models, Animal , Dysbiosis/drug therapy , Dysbiosis/microbiology , Dysbiosis/physiopathology , Inulin/administration & dosage , Male , Neuroglia/drug effects , Neuroglia/microbiology , Rats , Rats, Wistar
5.
Cell Mol Life Sci ; 75(7): 1145-1149, 2018 04.
Article in English | MEDLINE | ID: mdl-29285574

ABSTRACT

Post-infectious irritable bowel syndrome is a well-defined pathological entity that develops in about one-third of subjects after an acute infection (bacterial, viral) or parasitic infestation. Only recently it has been documented that an high incidence of post-infectious irritable bowel syndrome occurs after Clostridium difficile infection. However, until now it is not known why in some patients recovered from this infection the gastrointestinal disturbances persist for months or years. Based on our in vitro studies on enteric glial cells exposed to the effects of C. difficile toxin B, we hypothesize that persistence of symptoms up to the development of irritable bowel syndrome might be due to a disturbance/impairment of the correct functions of the enteroglial intestinal network.


Subject(s)
Clostridioides difficile/physiology , Clostridium Infections/microbiology , Enteric Nervous System/microbiology , Irritable Bowel Syndrome/microbiology , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridioides difficile/metabolism , Host-Pathogen Interactions , Humans , Intestinal Mucosa/innervation , Intestinal Mucosa/microbiology , Models, Theoretical , Neuroglia/microbiology , Risk Factors
6.
J Immunol ; 196(9): 3794-805, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26983788

ABSTRACT

Blood-brain barrier activation and/or dysfunction are a common feature of human neurobrucellosis, but the underlying pathogenic mechanisms are largely unknown. In this article, we describe an immune mechanism for inflammatory activation of human brain microvascular endothelial cells (HBMEC) in response to infection with Brucella abortus Infection of HBMEC with B. abortus induced the secretion of IL-6, IL-8, and MCP-1, and the upregulation of CD54 (ICAM-1), consistent with a state of activation. Culture supernatants (CS) from glial cells (astrocytes and microglia) infected with B. abortus also induced activation of HBMEC, but to a greater extent. Although B. abortus-infected glial cells secreted IL-1ß and TNF-α, activation of HBMEC was dependent on IL-1ß because CS from B. abortus-infected astrocytes and microglia deficient in caspase-1 and apoptosis-associated speck-like protein containing a CARD failed to induce HBMEC activation. Consistently, treatment of CS with neutralizing anti-IL-1ß inhibited HBMEC activation. Both absent in melanoma 2 and Nod-like receptor containing a pyrin domain 3 are partially required for caspase-1 activation and IL-1ß secretion, suggesting that multiple apoptosis-associated speck-like protein containing CARD-dependent inflammasomes contribute to IL-1ß-induced activation of the brain microvasculature. Inflammasome-mediated IL-1ß secretion in glial cells depends on TLR2 and MyD88 adapter-like/TIRAP. Finally, neutrophil and monocyte migration across HBMEC monolayers was increased by CS from Brucella-infected glial cells in an IL-1ß-dependent fashion, and the infiltration of neutrophils into the brain parenchyma upon intracranial injection of B. abortus was diminished in the absence of Nod-like receptor containing a pyrin domain 3 and absent in melanoma 2. Our results indicate that innate immunity of the CNS set in motion by B. abortus contributes to the activation of the blood-brain barrier in neurobrucellosis and IL-1ß mediates this phenomenon.


Subject(s)
Brain/immunology , Brucella abortus/immunology , Brucellosis/immunology , Neuroglia/immunology , Animals , Apoptosis Regulatory Proteins/metabolism , Blood-Brain Barrier/pathology , Brain/microbiology , CARD Signaling Adaptor Proteins , Cell Movement , Cells, Cultured , Female , Humans , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microvessels/pathology , Neuroglia/microbiology
7.
Cell Mol Life Sci ; 74(8): 1527-1551, 2017 04.
Article in English | MEDLINE | ID: mdl-27891552

ABSTRACT

Clostridium difficile causes nosocomial/antibiotic-associated diarrhoea and pseudomembranous colitis. The major virulence factors are toxin A and toxin B (TcdB), which inactivate GTPases by monoglucosylation, leading to cytopathic (cytoskeleton alteration, cell rounding) and cytotoxic effects (cell-cycle arrest, apoptosis). C. difficile toxins breaching the intestinal epithelial barrier can act on underlying cells, enterocytes, colonocytes, and enteric neurons, as described in vitro and in vivo, but until now no data have been available on enteric glial cell (EGC) susceptibility. EGCs are crucial for regulating the enteric nervous system, gut homeostasis, the immune and inflammatory responses, and digestive and extradigestive diseases. Therefore, we evaluated the effects of C. difficile TcdB in EGCs. Rat-transformed EGCs were treated with TcdB at 0.1-10 ng/ml for 1.5-48 h, and several parameters were analysed. TcdB induces the following in EGCs: (1) early cell rounding with Rac1 glucosylation; (2) early G2/M cell-cycle arrest by cyclin B1/Cdc2 complex inactivation caused by p27 upregulation, the downregulation of cyclin B1 and Cdc2 phosphorylated at Thr161 and Tyr15; and (3) apoptosis by a caspase-dependent but mitochondria-independent pathway. Most importantly, the stimulation of EGCs with TNF-α plus IFN-γ before, concomitantly or after TcdB treatment strongly increased TcdB-induced apoptosis. Furthermore, EGCs that survived the cytotoxic effect of TcdB did not recover completely and showed not only persistent Rac1 glucosylation, cell-cycle arrest and low apoptosis but also increased production of glial cell-derived neurotrophic factor, suggesting self-rescuing mechanisms. In conclusion, the high susceptibility of EGCs to TcdB in vitro, the increased sensitivity to inflammatory cytokines related to apoptosis and the persistence of altered functions in surviving cells suggest an important in vivo role of EGCs in the pathogenesis of C. difficile infection.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridioides difficile/physiology , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/pathology , Gastrointestinal Tract/innervation , Neuroglia/microbiology , Neuroglia/pathology , Animals , Apoptosis , Cell Cycle Checkpoints , Cell Line , Enterocolitis, Pseudomembranous/metabolism , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/pathology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Neuroglia/metabolism , Rats
8.
Cell Microbiol ; 17(10): 1464-76, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25864455

ABSTRACT

Borrelia burgdorferi, the agent of Lyme disease, spreads from the site of the tick bite to tissues such as heart, joints and the nervous tissues. Host glycosaminoglycans, highly modified repeating disaccharides that are present on cell surfaces and in extracellular matrix, are common targets of microbial pathogens during tissue colonization. While several dermatan sulfate-binding B. burgdorferi adhesins have been identified, B. burgdorferi adhesins documented to promote spirochetal binding to heparan sulfate have not yet been identified. OspEF-related proteins (Erps), a large family of plasmid-encoded surface lipoproteins that are produced in the mammalian host, can be divided into the OspF-related, OspEF-leader peptide (Elp) and OspE-related subfamilies. We show here that a member of the OspF-related subfamily, ErpG, binds to heparan sulfate and when produced on the surface of an otherwise non-adherent B. burgdorferi strain, ErpG promotes heparan sulfate-mediated bacterial attachment to the glial but not the endothelial, synovial or respiratory epithelial cells. Six other OspF-related proteins were capable of binding heparan sulfate, whereas representative OspE-related and Elp proteins lacked this activity. These results indicate that OspF-related proteins are heparan sulfate-binding adhesins, at least one of which promotes bacterial attachment to glial cells.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Adhesion , Bacterial Outer Membrane Proteins/metabolism , Borrelia burgdorferi/physiology , Glycosaminoglycans/metabolism , Heparitin Sulfate/metabolism , Lipoproteins/metabolism , Cells, Cultured , Neuroglia/microbiology , Protein Binding
9.
Immunol Cell Biol ; 93(9): 789-95, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25801479

ABSTRACT

Mycobacterium tuberculosis can infect 'non-classical immune cells', which comprise a significant constituency of cells that reside outside of those defined as 'classical immune cells' from myeloid or lymphoid origin. Here we address the influence of specific 'non-classical immune cells' in host responses and their effects in controlling mycobacterial growth or enabling an environment conducive for bacilli persistence. The interaction of M. tuberculosis with epithelial cells, endothelial cells, fibroblasts, adipocytes, glia and neurons and downstream cellular responses that often dictate immune regulation and disease outcome are discussed. Functional integration and synergy between 'classical' and 'non-classical immune cells' are highlighted as critical for determining optimal immune outcomes that favour the host.


Subject(s)
Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Adipocytes/immunology , Adipocytes/microbiology , Endothelial Cells/immunology , Endothelial Cells/microbiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Fibroblasts/immunology , Fibroblasts/microbiology , Humans , Models, Immunological , Mycobacterium tuberculosis/physiology , Neuroglia/immunology , Neuroglia/microbiology , Neurons/immunology , Neurons/microbiology , Tuberculosis/microbiology
10.
Gut ; 63(1): 105-15, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23292665

ABSTRACT

OBJECTIVE: Enteric glial cells (EGC) have been suggested to participate in host-bacteria cross-talk, playing a protective role within the gut. The way EGC interact with microorganisms is still poorly understood. We aimed to evaluate whether: EGC participate in host-bacteria interaction; S100B and Toll-like receptor (TLR) signalling converge in a common pathway leading to nitric oxide (NO) production. DESIGN: Primary cultures of human EGC were exposed to pathogenic (enteroinvasive Escherichia coli; EIEC) and probiotic (Lactobacillus paracasei F19) bacteria. Cell activation was assessed by evaluating the expression of cFos and major histocompatibility complex (MHC) class II molecules. TLR expression in EGC was evaluated at both baseline and after exposure to bacteria by real-time PCR, fluorescence microscopy and western blot analysis. S100B expression and NO release from EGC, following exposure to bacteria, were measured in the presence or absence of specific TLR and S100B pathway inhibitors. RESULTS: EIEC activated EGC by inducing the expression of cFos and MHC II. EGC expressed TLR at baseline. Pathogens and probiotics differentially modulated TLR expression in EGC. Pathogens, but not probiotics, significantly induced S100B protein overexpression and NO release from EGC. Pretreatment with specific inhibitors of TLR and S100B pathways abolished bacterial-induced NO release from EGC. CONCLUSIONS: Human EGC interact with bacteria and discriminate between pathogens and probiotics via a different TLR expression and NO production. In EGC, NO release is impaired in the presence of specific inhibitors of the TLR and S100B pathways, suggesting the presence of a novel common pathway involving both TLR stimulation and S100B protein upregulation.


Subject(s)
Escherichia coli/metabolism , Host-Pathogen Interactions , Intestine, Small/microbiology , Lactobacillus/metabolism , Neuroglia/microbiology , S100 Calcium Binding Protein beta Subunit/metabolism , Toll-Like Receptors/metabolism , Aged , Biomarkers/metabolism , Blotting, Western , Cells, Cultured , Female , Humans , Intestine, Small/metabolism , Male , Microscopy, Fluorescence , Middle Aged , Neuroglia/metabolism , Nitric Oxide/metabolism , Probiotics/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
11.
J Biol Chem ; 287(19): 15298-306, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22418440

ABSTRACT

Cryptococcus neoformans is a pathogenic yeast that can invade the brain and cause meningoencephalitis. Our previous in vitro studies suggested that the interaction between C. neoformans hyaluronic acid and human brain endothelial CD44 could be the initial step of brain invasion. In this report, we used a CD44 knock-out (KO or CD44(-/-)) mouse model to explore the importance of CD44 in C. neoformans brain invasion. Our results showed that C. neoformans-infected CD44 KO mice survived longer than the infected wild-type mice. Consistent with our in vitro results, the brain and cerebrospinal fluid fungal burden was reduced in CD44-deficient mice. Histopathological studies showed smaller and fewer cystic lesions in the brains of CD44 KO mice. Interestingly, the cystic lesions contained C. neoformans cells embedded within their polysaccharide capsule and were surrounded by host glial cells. We also found that a secondary hyaluronic acid receptor, RHAMM (receptor of hyaluronan-mediated motility), was present in the CD44 KO mice. Importantly, our studies demonstrated an in vivo blocking effect of simvastatin. These results suggest that the CD44 and RHAMM receptors function on membrane lipid rafts during invasion and that simvastatin may have a potential therapeutic role in C. neoformans infections of the brain.


Subject(s)
Brain/metabolism , Cryptococcosis/metabolism , Cryptococcus neoformans/metabolism , Extracellular Matrix Proteins/metabolism , Hyaluronan Receptors/metabolism , Animals , Blotting, Western , Brain/drug effects , Brain/microbiology , Cryptococcosis/cerebrospinal fluid , Cryptococcosis/microbiology , Cryptococcus neoformans/pathogenicity , Cryptococcus neoformans/physiology , Extracellular Matrix Proteins/genetics , Female , Host-Pathogen Interactions , Hyaluronan Receptors/genetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/microbiology , Protein Binding , RNA Interference , Simvastatin/pharmacology , Virulence
12.
J Immunol ; 186(12): 7089-97, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21602496

ABSTRACT

Muller cells, the principal glia of the retina, play several key roles in normal and various retinal diseases. To date, their direct involvement in retinal innate defense against bacterial pathogens has not been investigated. In this article, we show that Muller cells express TLR2, a key sensor implicated in recognizing Gram-positive bacteria. We found that intravitreal injection of TLR2 agonist Pam3Cys and Staphylococcus aureus activated Muller glia in C57BL/6 mouse retina. Similarly, Pam3Cys or S. aureus elicited the expression of TLR2 and activated the NF-κB and p38 MAPK signaling cascade. Concomitant with the activation of signaling pathways, transcriptional expression and secretion of various proinflammatory cytokines (IL-6, TNF-α, and IL-1ß), chemokines (IL-8), and antimicrobial peptide (LL-37) were also induced in Muller glia. Importantly, the culture media derived from TLR2-activated Muller glia exhibited robust bactericidal activity against S. aureus. Furthermore, use of neutralizing Ab, small interfering RNA, and pharmacological inhibitors revealed that Muller glial innate response to S. aureus is mediated via the TLR2-NF-κB axis. Collectively, this study for the first time, to our knowledge, establishes that the retinal Muller glia senses pathogens via TLR2 and contributes directly to retinal innate defense via production of inflammatory mediators and antimicrobial peptides.


Subject(s)
Neuroglia/microbiology , Retina/pathology , Staphylococcus aureus/immunology , Toll-Like Receptor 2/immunology , Animals , Cytokines , Immunity, Innate , Inflammation Mediators , Intravitreal Injections , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neuroglia/immunology , Retina/microbiology , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Inflammation ; 45(6): 2388-2405, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35776290

ABSTRACT

Enteric glial cells (EGCs) are involved in intestinal inflammation. In this study, we will investigate how Bifidobacterium bifidum (B.b.) and Bacteroides fragilis (B.f.) influence EGC regulation. After pretreatment with lipopolysaccharide (LPS) and interferon-γ (IFN-γ), the expressions of major histocompatibility complex class II (MHC-II), CD80, CD86, glial cell line-derived neurotrophic factor (GDNF), toll-like receptor 2 (TLR-2), and tumor necrosis factor-α (TNF-α) in EGCs were detected using polymerase chain reaction and western blot after co-culture with the supernatants of B.b. or B.f. (multiplicity of infection, 40:1 or 80:1). Finally, EGCs were co-cultured with naive CD4+ T cells, and the expressions of interleukin (IL)-2, IL-4, IL-10, and IL-17 in supernatant were measured using enzyme-linked immunosorbent assay (ELISA). The mRNA expressions of MHC-II and CD86 in EGCs were increased after combined stimulation with LPS and IFN-γ. The expressions of MHC-II, GDNF, TLR-2, and TNF-α were all significantly upregulated in stimulated EGCs. The B.b. supernatant downregulated the expressions of MHC-II, GDNF, TLR-2, and TNF-α in stimulated EGCs, whereas the B.f. supernatant upregulated TLR-2 expression and downregulated MHC-II expression. The expressions of IL-4, IL-2, and IL-17 after co-culture of naive CD4+ T cells and stimulated EGCs were significantly increased. The supernatant of B.b. or B.f. downregulated the expressions of these cytokines. The low-concentration B.b. supernatant upregulated IL-10 expression. Conclusions B.b. and B.f. may influence intestinal inflammation by regulating MHC-II, GDNF, TLR-2, and TNF-α expression in EGCs and IL-4, IL-2, IL-17, and IL-10 secretion.


Subject(s)
Bacteroides fragilis , Bifidobacterium bifidum , Neuroglia , Humans , Bacteroides fragilis/metabolism , Bifidobacterium bifidum/metabolism , Cells, Cultured , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Inflammation/metabolism , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-17/metabolism , Interleukin-2 , Interleukin-4/metabolism , Lipopolysaccharides , Neuroglia/metabolism , Neuroglia/microbiology , Toll-Like Receptor 2/metabolism , Tumor Necrosis Factor-alpha/metabolism
14.
Sci Rep ; 11(1): 10722, 2021 05 21.
Article in English | MEDLINE | ID: mdl-34021227

ABSTRACT

Staphylococcus aureus infections of the central nervous system are serious and can be fatal. S. aureus is commonly present in the nasal cavity, and after injury to the nasal epithelium it can rapidly invade the brain via the olfactory nerve. The trigeminal nerve constitutes another potential route of brain infection. The glia of these nerves, olfactory ensheathing cells (OECs) and trigeminal nerve Schwann cells (TgSCs), as well as astrocytes populating the glia limitans layer, can phagocytose bacteria. Whilst some glial responses to S. aureus have been studied, the specific responses of different glial types are unknown. Here, we compared how primary mouse OECs, TgSCs, astrocytes and microglia responded to S. aureus. All glial types internalized the bacteria within phagolysosomes, and S. aureus-conjugated BioParticles could be tracked with subtle but significant differences in time-course of phagocytosis between glial types. Live bacteria could be isolated from all glia after 24 h in culture, and microglia, OECs and TgSCs exhibited better protection against intracellular S. aureus survival than astrocytes. All glial types responded to the bacteria by cytokine secretion. Overall, OECs secreted the lowest level of cytokines, suggesting that these cells, despite showing strong capacity for phagocytosis, have immunomodulatory functions that can be relevant for neural repair.


Subject(s)
Central Nervous System/microbiology , Disease Resistance , Host-Pathogen Interactions , Neuroglia/microbiology , Peripheral Nervous System/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Biomarkers , Cells, Cultured , Central Nervous System/immunology , Cytokines/metabolism , Disease Resistance/immunology , Host-Pathogen Interactions/immunology , Microglia , Neuroglia/immunology , Neuroglia/metabolism , Peripheral Nervous System/immunology , Phagocytosis/immunology , Staphylococcal Infections/immunology
15.
Cell Rep ; 37(13): 110166, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34965433

ABSTRACT

Animals encounter microorganisms in their habitats, adapting physiology and behavior accordingly. The nematode Caenorhabditis elegans is found in microbe-rich environments; however, its responses to fungi are not extensively studied. Here, we describe interactions of C. elegans and Penicillium brevicompactum, an ecologically relevant mold. Transcriptome studies reveal that co-culture upregulates stress response genes, including xenobiotic-metabolizing enzymes (XMEs), in C. elegans intestine and AMsh glial cells. The nuclear hormone receptors (NHRs) NHR-45 and NHR-156 are induction regulators, and mutants that cannot induce XMEs in the intestine when exposed to P. brevicompactum experience mitochondrial stress and exhibit developmental defects. Different C. elegans wild isolates harbor sequence polymorphisms in nhr-156, resulting in phenotypic diversity in AMsh glia responses to microbe exposure. We propose that P. brevicompactum mitochondria-targeting mycotoxins are deactivated by intestinal detoxification, allowing tolerance to moldy environments. Our studies support the idea that C. elegans NHRs may be regulated by environmental cues.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/drug effects , Gastrointestinal Tract/enzymology , Mitochondria/enzymology , Neuroglia/enzymology , Penicillium/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/genetics , Enzyme Induction , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/microbiology , Gene Expression Regulation, Developmental , Mitochondria/drug effects , Mitochondria/microbiology , Neuroglia/drug effects , Neuroglia/microbiology
16.
Arkh Patol ; 72(2): 36-7, 2010.
Article in Russian | MEDLINE | ID: mdl-20698315

ABSTRACT

The authors describe a case of Lyme disease--neuroborreliosis. In neuroborreliosis, there are morphohistological changes: pronounced dystrophic processes in the brain nerve cells, spongiosis, perivascular hemorrhagic infiltrations, glial proliferation with the formation of perivascular glial granulomas.


Subject(s)
Brain/pathology , Granuloma/pathology , Intracranial Hemorrhages/pathology , Lyme Neuroborreliosis/pathology , Neuroglia/pathology , Aged , Brain/microbiology , Fatal Outcome , Granuloma/microbiology , Humans , Intracranial Hemorrhages/microbiology , Male , Neuroglia/microbiology
17.
J Neuroinflammation ; 6: 23, 2009 Aug 25.
Article in English | MEDLINE | ID: mdl-19706181

ABSTRACT

BACKGROUND: Lyme neuroborreliosis (LNB) may present as meningitis, cranial neuropathy, acute radiculoneuropathy or, rarely, as encephalomyelitis. We hypothesized that glia, upon exposure to Borrelia burgdorferi, the Lyme disease agent, produce inflammatory mediators that promote the acute cellular infiltration of early LNB. This inflammatory context could potentiate glial and neuronal apoptosis. METHODS: We inoculated live B. burgdorferi into the cisterna magna of rhesus macaques and examined the inflammatory changes induced in the central nervous system (CNS), and dorsal root nerves and ganglia (DRG). RESULTS: ELISA of the cerebrospinal fluid (CSF) showed elevated IL-6, IL-8, CCL2, and CXCL13 as early as one week post-inoculation, accompanied by primarily lymphocytic and monocytic pleocytosis. In contrast, onset of the acquired immune response, evidenced by anti-B. burgdorferi C6 serum antibodies, was first detectable after 3 weeks post-inoculation. CSF cell pellets and CNS tissues were culture-positive for B. burgdorferi. Histopathology revealed signs of acute LNB: severe multifocal leptomeningitis, radiculitis, and DRG inflammatory lesions. Immunofluorescence staining and confocal microscopy detected B. burgdorferi antigen in the CNS and DRG. IL-6 was observed in astrocytes and neurons in the spinal cord, and in neurons in the DRG of infected animals. CCL2 and CXCL13 were found in microglia as well as in endothelial cells, macrophages and T cells. Importantly, the DRG of infected animals showed significant satellite cell and neuronal apoptosis. CONCLUSION: Our results support the notion that innate responses of glia to B. burgdorferi initiate/mediate the inflammation seen in acute LNB, and show that neuronal apoptosis occurs in this context.


Subject(s)
Encephalitis/physiopathology , Lyme Neuroborreliosis/physiopathology , Meningitis/physiopathology , Neuroglia/immunology , Radiculopathy/physiopathology , Spinal Cord Diseases/physiopathology , Animals , Antibodies/blood , Apoptosis/immunology , Brain/immunology , Brain/pathology , Brain/physiopathology , Chemokines/metabolism , Encephalitis/immunology , Encephalitis/microbiology , Ganglia, Spinal/immunology , Ganglia, Spinal/pathology , Ganglia, Spinal/physiopathology , Gliosis/immunology , Gliosis/microbiology , Gliosis/physiopathology , Leukocytosis/immunology , Leukocytosis/microbiology , Leukocytosis/physiopathology , Lyme Neuroborreliosis/immunology , Lyme Neuroborreliosis/pathology , Macaca mulatta , Meningitis/immunology , Meningitis/microbiology , Nerve Degeneration/immunology , Nerve Degeneration/microbiology , Nerve Degeneration/physiopathology , Neuroglia/microbiology , Neurons/immunology , Neurons/microbiology , Neurons/pathology , Radiculopathy/immunology , Radiculopathy/microbiology , Spinal Cord/immunology , Spinal Cord/pathology , Spinal Cord/physiopathology , Spinal Cord Diseases/immunology , Spinal Cord Diseases/microbiology
18.
Am J Pathol ; 173(5): 1415-27, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18832582

ABSTRACT

Lyme neuroborreliosis, caused by the spirochete Borrelia burgdorferi, often manifests by causing neurocognitive deficits. As a possible mechanism for Lyme neuroborreliosis, we hypothesized that B. burgdorferi induces the production of inflammatory mediators in the central nervous system with concomitant neuronal and/or glial apoptosis. To test our hypothesis, we constructed an ex vivo model that consisted of freshly collected slices from brain cortex of a rhesus macaque and allowed live B. burgdorferi to penetrate the tissue. Numerous transcripts of genes that regulate inflammation as well as oligodendrocyte and neuronal apoptosis were significantly altered as assessed by DNA microarray analysis. Transcription level increases of 7.43-fold (P = 0.005) for the cytokine tumor necrosis factor-alpha and 2.31-fold (P = 0.016) for the chemokine interleukin (IL)-8 were also detected by real-time-polymerase chain reaction array analysis. The immune mediators IL-6, IL-8, IL-1beta, COX-2, and CXCL13 were visualized in glial cells in situ by immunofluorescence staining and confocal microscopy. Concomitantly, significant proportions of both oligodendrocytes and neurons undergoing apoptosis were present in spirochete-stimulated tissues. IL-6 production by astrocytes in addition to oligodendrocyte apoptosis were also detected, albeit at lower levels, in rhesus macaques that had received in vivo intraparenchymal stereotaxic inoculations of live B. burgdorferi. These results provide proof of concept for our hypothesis that B. burgdorferi produces inflammatory mediators in the central nervous system, accompanied by glial and neuronal apoptosis.


Subject(s)
Apoptosis , Borrelia burgdorferi/physiology , Brain/microbiology , Inflammation Mediators/metabolism , Lyme Disease/microbiology , Neuroglia/pathology , Neurons/pathology , Animals , Apoptosis/genetics , Brain/immunology , Brain/pathology , Cytokines/biosynthesis , Female , Gene Expression Regulation , Humans , Immune System , Lyme Disease/immunology , Macaca mulatta , Male , Microscopy, Confocal , Neuroglia/immunology , Neuroglia/microbiology , Neurons/microbiology , Oligodendroglia/pathology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
19.
Science ; 226(4680): 1337-9, 1984 Dec 14.
Article in English | MEDLINE | ID: mdl-6095453

ABSTRACT

A human papovavirus, JCV, is the etiologic agent of the fatal demyelinating disease, progressive multifocal leukoencephalopathy. The JCV 98-base-pair tandem repeats, located to the late side of the viral replication origin, were shown to be a transcriptional regulatory element with enhancer-like activity in human fetal glial cells. These tandem repeats share significant homology with the 82-nucleotide rat brain-specific identifier RNA sequence.


Subject(s)
Brain/microbiology , JC Virus/genetics , Operon , Polyomavirus/genetics , Base Sequence , Fetus , Gene Amplification , Gene Expression Regulation , Genes, Viral , Humans , Neuroglia/microbiology
20.
Science ; 159(3822): 1476-7, 1968 Mar 29.
Article in English | MEDLINE | ID: mdl-4307975

ABSTRACT

Astrocytes in cultures of brain cells from fetal or newborn hamsters undergo neoplastic transformation after infection with simian virus 40 or polyoma virus. Subcutaneous or intracerebral inoculation of the transformed brain cells into newborn or adult hamsters produces progressively enlarging astrocytomas at the sites of injection. Astrocytomas produced by polyomatransformed cell lines are histologically better differentiated, but grow more rapidly and metastasize more frequently, than astrocytomas produced by cell lines transformed by simian virus 40. These observations make available in vitro models of virus-induced oncogenesis in astrocytes and provide simple techniques for obtaining astrocytoma cell lines suitable for screening studies of chemical agents effective against astrocytomas.


Subject(s)
Astrocytoma/etiology , Cytopathogenic Effect, Viral , Neuroglia/microbiology , Polyomavirus/pathogenicity , Simian virus 40/pathogenicity , Animals , Animals, Newborn , Cell Differentiation , Cell Transformation, Neoplastic , Cricetinae , Culture Techniques , Fetus , Neoplasm Metastasis
SELECTION OF CITATIONS
SEARCH DETAIL