Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 214
Filtrar
1.
Inflammation ; 45(6): 2388-2405, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35776290

RESUMO

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.


Assuntos
Bacteroides fragilis , Bifidobacterium bifidum , Neuroglia , Humanos , Bacteroides fragilis/metabolismo , Bifidobacterium bifidum/metabolismo , Células Cultivadas , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Inflamação/metabolismo , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Interleucina-2 , Interleucina-4/metabolismo , Lipopolissacarídeos , Neuroglia/metabolismo , Neuroglia/microbiologia , Receptor 2 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
2.
Cell Rep ; 37(13): 110166, 2021 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-34965433

RESUMO

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.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/efeitos dos fármacos , Trato Gastrointestinal/enzimologia , Mitocôndrias/enzimologia , Neuroglia/enzimologia , Penicillium/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/microbiologia , Proteínas de Caenorhabditis elegans/genética , Indução Enzimática , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Regulação da Expressão Gênica no Desenvolvimento , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/microbiologia , Neuroglia/efeitos dos fármacos , Neuroglia/microbiologia
3.
Sci Rep ; 11(1): 10722, 2021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-34021227

RESUMO

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.


Assuntos
Sistema Nervoso Central/microbiologia , Resistência à Doença , Interações Hospedeiro-Patógeno , Neuroglia/microbiologia , Sistema Nervoso Periférico/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/fisiologia , Biomarcadores , Células Cultivadas , Sistema Nervoso Central/imunologia , Citocinas/metabolismo , Resistência à Doença/imunologia , Interações Hospedeiro-Patógeno/imunologia , Microglia , Neuroglia/imunologia , Neuroglia/metabolismo , Sistema Nervoso Periférico/imunologia , Fagocitose/imunologia , Infecções Estafilocócicas/imunologia
4.
Neurosci Lett ; 708: 134334, 2019 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-31238130

RESUMO

Glia are key regulators of inflammatory responses within the central nervous system (CNS) following infection or trauma. We have previously demonstrated the ability of activated astrocytes to rapidly produce pro-inflammatory mediators followed by a transition to an anti-inflammatory cytokine production profile that includes the immunosuppressive cytokine interleukin (IL)-10 and the closely related cytokines IL-19 and IL-24. IL-20, another member of the IL-10 family, is known to modulate immune cell activity in the periphery and we have previously demonstrated that astrocytes constitutively express the cognate receptors for this cytokine. However, the ability of glia to produce IL-20 remains unclear and the effects of this pleiotropic cytokine on glial immune functions have not been investigated. In this study, we report that primary murine and human astrocytes are not an appreciable source of IL-20 following challenge with disparate bacterial species or their components. Importantly, we have determined that astrocyte are responsive to the immunomodulatory actions of this cytokine by showing that recombinant IL-20 administration upregulates microbial pattern recognition receptor expression and induces release of the inflammatory mediator IL-6 by these cells. Taken together, these data suggest that IL-20 acts in a dissimilar manner to other IL-10 family members to augment the inflammatory responses of astrocytes.


Assuntos
Astrócitos/metabolismo , Interleucinas/metabolismo , Animais , Astrócitos/microbiologia , Células Cultivadas , Humanos , Imunomodulação , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Interleucinas/farmacologia , Camundongos Endogâmicos C57BL , Neisseria meningitidis/fisiologia , Neuroglia/metabolismo , Neuroglia/microbiologia , Proteínas Recombinantes/farmacologia , Staphylococcus aureus/fisiologia , Streptococcus pneumoniae/fisiologia , Receptores Toll-Like/metabolismo
5.
J Neuroinflammation ; 16(1): 113, 2019 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-31138231

RESUMO

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.


Assuntos
Edema/patologia , Hiperalgesia/patologia , Leishmaniose/patologia , Neuroglia/patologia , Dor/patologia , Medula Espinal/patologia , Animais , Edema/microbiologia , Hiperalgesia/microbiologia , Leishmania , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neuroglia/microbiologia , Dor/microbiologia , Medula Espinal/microbiologia
6.
Eur J Nutr ; 58(8): 3361-3375, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-30826905

RESUMO

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.


Assuntos
Ansiolíticos/farmacologia , Anti-Inflamatórios/farmacologia , Encéfalo/efeitos dos fármacos , Diabetes Mellitus Experimental/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Inulina/farmacologia , Lactobacillus plantarum , Simbióticos/administração & dosagem , Animais , Ansiolíticos/administração & dosagem , Anti-Inflamatórios/administração & dosagem , Encéfalo/microbiologia , Diabetes Mellitus Experimental/fisiopatologia , Modelos Animais de Doenças , Disbiose/tratamento farmacológico , Disbiose/microbiologia , Disbiose/fisiopatologia , Inulina/administração & dosagem , Masculino , Neuroglia/efeitos dos fármacos , Neuroglia/microbiologia , Ratos , Ratos Wistar
7.
J Biomed Sci ; 26(1): 16, 2019 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-30732602

RESUMO

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.


Assuntos
Edema/fisiopatologia , Escherichia coli Êntero-Hemorrágica/fisiologia , Hipocampo/fisiopatologia , Peroxidação de Lipídeos , Lipopolissacarídeos/efeitos adversos , Toxina Shiga II/efeitos adversos , Animais , Edema/microbiologia , Hipocampo/efeitos dos fármacos , Hipocampo/microbiologia , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Neuroglia/efeitos dos fármacos , Neuroglia/microbiologia , Neuroglia/fisiologia
8.
Biochim Biophys Acta Mol Cell Res ; 1865(12): 1945-1958, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30296499

RESUMO

Clostridium difficile infection (CDI) causes nosocomial/antibiotic-associated diarrhea and pseudomembranous colitis, with dramatic incidence/mortality worldwide. C. difficile virulence factors are toxin A and toxin B (TcdB) which cause cytopathic/cytotoxic effects and inflammation. Until now studies were focused on molecular effects of C. difficile toxins (Tcds) on different cells while unexplored aspect is the status/fate of cells that survived their cytotoxicity. Recently we demonstrated that enteric glial cells (EGCs) are susceptible to TcdB cytotoxicity, but several EGCs survived and were irreversibly cell-cycle arrested and metabolically active, suggesting that EGCs could became senescent. This is important because allowed us to evaluate the not explored status/fate of cells surviving Tcds cytotoxicity, and particularly if TcdB induces senescence in EGCs. Rat-transformed EGCs were treated with 10 ng/ml TcdB for 6 h-48 h, or for 48 h, followed by incubation for additional 4 or 11 days in absence of TcdB (6 or 13 total days). Senescence markers/effectors were examined by specific assays. TcdB induces senescence in EGCs, as demonstrated by the senescence markers: irreversible cell-cycle arrest, senescence-associated-ß­galactosidase positivity, flat morphology, early and persistent DNA damage (ATM and H2AX phosphorylation), p27 overexpression, pRB hypophosphorylation, c­Myc, cyclin B1, cdc2 and phosphorylated-cdc2 downregulation, Sirtuin­2 and Sirtuin­3 overexpression. TcdB-induced EGC senescence is dependent by JNK and AKT activation but independent by ROS, p16 and p53/p21 pathways. In conclusion, TcdB induces senescence in EGCs. The extrapolation of these results to CDI leads to hypothesize that EGCs that survived TcdB, once they have acquired a senescence state, could cause irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), and tumors due to persistent inflammation, transfer of senescence status and stimulation of pre-neoplastic cells.


Assuntos
Proteínas de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Clostridioides difficile/patogenicidade , Neuroglia/citologia , Animais , Pontos de Checagem do Ciclo Celular , Células Cultivadas , Senescência Celular , Clostridioides difficile/metabolismo , Dano ao DNA , Regulação da Expressão Gênica/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Neuroglia/microbiologia , Ratos , Transdução de Sinais
9.
PLoS One ; 13(5): e0197413, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29746581

RESUMO

Borrelia burgdorferi, the agent of Lyme borreliosis, can elude hosts' innate and adaptive immunity as part of the course of infection. The ability of B. burgdorferi to invade or be internalized by host cells in vitro has been proposed as a mechanism for the pathogen to evade immune responses or antimicrobials. We have previously shown that B. burgdorferi can be internalized by human neuroglial cells. In this study we demonstrate that these cells take up B. burgdorferi via coiling phagocytosis mediated by the formin, Daam1, a process similarly described for human macrophages. Following coincubation with glial cells, B. burgdorferi was enwrapped by Daam1-enriched coiling pseudopods. Coiling of B. burgdorferi was significantly reduced when neuroglial cells were pretreated with anti-Daam1 antibody indicating the requirement for Daam1 for borrelial phagocytosis. Confocal microscopy showed Daam1 colocalizing to the B. burgdorferi surface suggesting interaction with borrelial membrane protein(s). Using the yeast 2-hybrid system for identifying protein-protein binding, we found that the B. burgdorferi surface lipoprotein, BBA66, bound the FH2 subunit domain of Daam1. Recombinant proteins were used to validate binding by ELISA, pull-down, and co-immunoprecipitation. Evidence for native Daam1 and BBA66 interaction was suggested by colocalization of the proteins in the course of borrelial capture by the Daam1-enriched pseudopodia. Additionally, we found a striking reduction in coiling for a BBA66-deficient mutant strain compared to BBA66-expressing strains. These results show that coiling phagocytosis is a mechanism for borrelial internalization by neuroglial cells mediated by Daam1.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Borrelia burgdorferi , Doença de Lyme/imunologia , Neurônios/microbiologia , Neutrófilos/metabolismo , Fagocitose , Imunidade Adaptativa , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioma/metabolismo , Glioma/patologia , Humanos , Imunidade Inata , Lipoproteínas/química , Macrófagos/metabolismo , Proteínas dos Microfilamentos , Neuroglia/metabolismo , Neuroglia/microbiologia , Ligação Proteica , Domínios Proteicos , Proteínas Recombinantes/química , Técnicas do Sistema de Duplo-Híbrido , Proteínas rho de Ligação ao GTP
10.
Cell Mol Life Sci ; 75(7): 1145-1149, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29285574

RESUMO

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.


Assuntos
Clostridioides difficile/fisiologia , Infecções por Clostridium/microbiologia , Sistema Nervoso Entérico/microbiologia , Síndrome do Intestino Irritável/microbiologia , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Mucosa Intestinal/inervação , Mucosa Intestinal/microbiologia , Modelos Teóricos , Neuroglia/microbiologia , Fatores de Risco
11.
Free Radic Biol Med ; 112: 608-615, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28916475

RESUMO

NADH oxidase (NOX) plays important roles in respiration and reactive oxygen species (ROS) generation in cells. In this study, we explored the function of NOX in Listeria monocytogenes by gene deletion. From our results, nox mutant strain (∆nox) had lower H2O2 level and showed no significant alteration in bacteria growth activity. But it had enhanced invasiveness during the invasion of glial cells and mice brain compared to wildtype strain. Furthermore, several virulence genes involved in invasion, such as inlA, inlB, vip and sigB, were upregulated in ∆nox, and the alterations could be restored by complementation. To explore if nox was involved in the interaction of pathogen and host, we examined the generation of host ROS including superoxide and H2O2 during infection, and found ∆nox invasion leading to less superoxide and H2O2 generation. Besides, the upregulation of pro-inflammatory factors in glial cells was restrained when invaded by ∆nox compared to wildtype and complementary strain. In conclusion, our study evaluated the function of nox in L. monocytogenes and indicated that nox could regulate the invasion of L. monocytogenes by regulating virulence genes expression and the interaction of host-and- pathogens.


Assuntos
Sequência de Bases , Regulação Bacteriana da Expressão Gênica , Listeria monocytogenes/genética , Listeriose/genética , NADPH Oxidases/genética , Deleção de Sequência , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Encéfalo/metabolismo , Encéfalo/microbiologia , Encéfalo/patologia , Teste de Complementação Genética , Interações Hospedeiro-Patógeno , Peróxido de Hidrogênio/metabolismo , Listeria monocytogenes/crescimento & desenvolvimento , Listeria monocytogenes/patogenicidade , Listeriose/metabolismo , Listeriose/microbiologia , Listeriose/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , NADPH Oxidases/deficiência , Neuroglia/metabolismo , Neuroglia/microbiologia , Neuroglia/patologia , Fator sigma/genética , Fator sigma/metabolismo , Transdução de Sinais , Superóxidos/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
12.
Br J Pharmacol ; 174(23): 4295-4307, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28888095

RESUMO

BACKGROUND AND PURPOSE: Streptococcus pneumoniae is the most common cause of bacterial meningitis in adults and is characterized by high lethality and substantial cognitive disabilities in survivors. Here, we have studied the capacity of an established therapeutic agent, magnesium, to improve survival in pneumococcal meningitis by modulating the neurological effects of the major pneumococcal pathogenic factor, pneumolysin. EXPERIMENTAL APPROACH: We used mixed primary glial and acute brain slice cultures, pneumolysin injection in infant rats, a mouse meningitis model and complementary approaches such as Western blot, a black lipid bilayer conductance assay and live imaging of primary glial cells. KEY RESULTS: Treatment with therapeutic concentrations of magnesium chloride (500 mg·kg-1 in animals and 2 mM in cultures) prevented pneumolysin-induced brain swelling and tissue remodelling both in brain slices and in animal models. In contrast to other divalent ions, which diminish the membrane binding of pneumolysin in non-therapeutic concentrations, magnesium delayed toxin-driven pore formation without affecting its membrane binding or the conductance profile of its pores. Finally, magnesium prolonged the survival and improved clinical condition of mice with pneumococcal meningitis, in the absence of antibiotic treatment. CONCLUSIONS AND IMPLICATIONS: Magnesium is a well-established and safe therapeutic agent that has demonstrated capacity for attenuating pneumolysin-triggered pathogenic effects on the brain. The improved animal survival and clinical condition in the meningitis model identifies magnesium as a promising candidate for adjunctive treatment of pneumococcal meningitis, together with antibiotic therapy.


Assuntos
Cloreto de Magnésio/administração & dosagem , Meningite Pneumocócica/tratamento farmacológico , Streptococcus pneumoniae/efeitos dos fármacos , Estreptolisinas/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/microbiologia , Modelos Animais de Doenças , Feminino , Cloreto de Magnésio/farmacologia , Meningite Pneumocócica/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/efeitos dos fármacos , Neuroglia/microbiologia , Ratos , Ratos Sprague-Dawley , Streptococcus pneumoniae/isolamento & purificação , Taxa de Sobrevida
13.
Cell Mol Life Sci ; 74(8): 1527-1551, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27891552

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/fisiologia , Enterocolite Pseudomembranosa/microbiologia , Enterocolite Pseudomembranosa/patologia , Trato Gastrointestinal/inervação , Neuroglia/microbiologia , Neuroglia/patologia , Animais , Apoptose , Pontos de Checagem do Ciclo Celular , Linhagem Celular , Enterocolite Pseudomembranosa/metabolismo , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Neuroglia/metabolismo , Ratos
14.
Immunity ; 45(2): 238-9, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27533011

RESUMO

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.


Assuntos
Disbiose/genética , Microbioma Gastrointestinal/imunologia , Imunidade Inata , Intestinos/imunologia , Linfócitos/imunologia , Neuroglia/imunologia , Proteínas Proto-Oncogênicas c-ret/metabolismo , Animais , Técnicas de Silenciamento de Genes , Fatores Neurotróficos Derivados de Linhagem de Célula Glial/metabolismo , Homeostase , Humanos , Interleucinas/metabolismo , Intestinos/inervação , Camundongos , Neuroglia/microbiologia , Proteínas Proto-Oncogênicas c-ret/genética , Simbiose , Interleucina 22
15.
J Immunol ; 196(9): 3794-805, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26983788

RESUMO

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.


Assuntos
Encéfalo/imunologia , Brucella abortus/imunologia , Brucelose/imunologia , Neuroglia/imunologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Barreira Hematoencefálica/patologia , Encéfalo/microbiologia , Proteínas Adaptadoras de Sinalização CARD , Movimento Celular , Células Cultivadas , Feminino , Humanos , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microvasos/patologia , Neuroglia/microbiologia
16.
Neuroscience ; 317: 149-61, 2016 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-26791522

RESUMO

Streptococcus pneumoniae is the causative agent of numerous diseases including severe invasive infections such as bacteremia and meningitis. It has been previously shown that strains of S. pneumoniae that are unable to survive in the bloodstream may colonize the CNS. However, information on cellular components and pathways involved in the neurotropism of these strains is still scarce. The olfactory system is a specialized tissue in which olfactory receptor neurons (ORNs) are interfacing with the external environment through several microvilli. Olfactory ensheathing cells (OECs) which also form the glial limiting membrane at the surface of the olfactory bulb (OB) are the only cells that ensheathe the ORNs axons. Since previous data from our group showed that OECs may harbor S. pneumoniae, we decided to test whether infection of the OB or OEC cultures modulates the expression levels of neurotrophic factor's mRNA and its putative effects on the activation and viability of microglia. We observed that neurotrophin-3 (NT-3) and glial cell-line-derived neurotrophic factor (GDNF) expression was significantly higher in the OB from uninfected mice than in infected mice. A similar result was observed when we infected OEC cultures. Brain-derived neurotrophic factor (BNDF) expression was significantly lower in the OB from infected mice than in uninfected mice. In contrast, in vitro infection of OECs resulted in a significant increase of BDNF mRNA expression. An upregulation of high-mobility group box 1 (HMGB1) expression was observed in both OB and OEC cultures infected with S. pneumoniae. Moreover, we found that conditioned medium from infected OEC cultures induced the expression of the pro-apoptotic protein cleaved-caspase-3 and an apparently continuous nuclear factor-kappa B (NF-κB) p65 activation in the N13 microglia. Altogether, our data suggest the possible existence of an OEC-pathogen molecular interface, through which the OECs could interfere on the activation and viability of microglia, favoring the access of non-hematogenous S. pneumoniae strains to the CNS in the absence of bacteremia.


Assuntos
Fatores de Crescimento Neural/metabolismo , Neuroglia/metabolismo , Bulbo Olfatório/metabolismo , Bulbo Olfatório/patologia , Infecções Pneumocócicas/patologia , 2',3'-Nucleotídeo Cíclico Fosfodiesterases/metabolismo , Actinas/metabolismo , Animais , Caspase 3/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Regulação Bacteriana da Expressão Gênica/fisiologia , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , N-Acetil-Muramil-L-Alanina Amidase/genética , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , NF-kappa B/metabolismo , Fatores de Crescimento Neural/genética , Neuroglia/microbiologia , RNA Mensageiro/metabolismo
17.
FEMS Microbiol Lett ; 363(2): fnv228, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26635415

RESUMO

Listeria monocytogenes is a foodborne pathogen that could cause severe infection in the central nervous system of humans and animals. However, the molecular mechanism of the pathogenesis is not fundamentally assessed. This study aimed to analyze the role of reactive oxygen species (ROS) in L. monocytogenes during its invasion into glia cells. The ROS level in L. monocytogenes was manipulated using NAD(P)H oxidase inhibitor diphenyleneiodonium chloride (DPI) and ROS scavenger N-acetyl cysteine (NAC). Results showed that the invasiveness of L. monocytogenes was elevated when ROS was downregulated by DPI and NAC treatment. Expression profiles of proinflammatory factors in glia cells were also examined because they play important roles in the functions of glia cells in the brain immune system. The expression levels of proinflammatory factors (tumor necrosis factor α and interleukin-1ß) in host glia cells were downregulated when invaded by L. monocytogenes with lower ROS level. This finding indicates that ROS may function as negative regulator during the invasion of L. monocytogenes in brain infection.


Assuntos
Listeria monocytogenes/fisiologia , Listeriose/metabolismo , Neuroglia/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular , Interações Hospedeiro-Patógeno , Humanos , Listeria monocytogenes/genética , Listeriose/microbiologia , Neuroglia/microbiologia
18.
Insect Sci ; 23(2): 253-64, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25394184

RESUMO

The pathogenic Wolbachia strain wMelPop rapidly over-replicates in the brain, muscles, and retina of Drosophila melanogaster, causing severe tissue degeneration and premature death of the host. The unique features of this endosymbiont make it an excellent tool to be used for biological control of insects, pests, and vectors of human diseases. To follow the dynamics of bacterial morphology and titer in the nerve cells we used transmission electron microscopy of 3-d-old female brains. The neurons and glial cells from central brain of the fly had different Wolbachia titers ranging from single bacteria to large accumulations, tearing cell apart and invading extracellular space. The neuropile regions of the brain were free of wMelPop. Wolbachia tightly interacted with host cell organelles and underwent several morphological changes in nerve cells. Based on different morphological types of bacteria described we propose for the first time a scheme of wMelPop dynamics within the somatic tissue of the host.


Assuntos
Drosophila melanogaster/microbiologia , Wolbachia/fisiologia , Animais , Encéfalo/microbiologia , Encéfalo/ultraestrutura , Drosophila melanogaster/ultraestrutura , Feminino , Microscopia Eletrônica de Transmissão , Neuroglia/microbiologia , Neurônios/microbiologia
19.
Curr Protoc Neurosci ; 72: 2.23.1-2.23.14, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26131661

RESUMO

In the central nervous system (CNS), myelin is formed by oligodendrocytes that are derived from precursor cells, known as oligodendrocyte precursor cells (OPCs). Successive stages of OPC interactions with the axons can be visualized in vitro and ex vivo using mixed neural cell cultures and pieces of intact spinal cord, respectively. OPCs and their differentiation can be imaged using cell-type-specific markers or green fluorescent protein (GFP) tags. This protocol describes methodology for generating these two systems for time-lapse imaging of dynamic cell interactions using fluorescent and 2-photon microscopy.


Assuntos
Axônios/fisiologia , Sistema Nervoso Central/citologia , Neuroglia/citologia , Imagem com Lapso de Tempo/métodos , Animais , Corantes Fluorescentes/metabolismo , Neuroglia/microbiologia
20.
Cell Microbiol ; 17(10): 1464-76, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25864455

RESUMO

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.


Assuntos
Antígenos de Bactérias/metabolismo , Aderência Bacteriana , Proteínas da Membrana Bacteriana Externa/metabolismo , Borrelia burgdorferi/fisiologia , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/metabolismo , Lipoproteínas/metabolismo , Células Cultivadas , Neuroglia/microbiologia , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA