Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 557
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 115(25): 6458-6463, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29866843

RESUMO

The enteric nervous system (ENS) is crucial for essential gastrointestinal physiologic functions such as motility, fluid secretion, and blood flow. The gut is colonized by trillions of bacteria that regulate host production of several signaling molecules including serotonin (5-HT) and other hormones and neurotransmitters. Approximately 90% of 5-HT originates from the intestine, and activation of the 5-HT4 receptor in the ENS has been linked to adult neurogenesis and neuroprotection. Here, we tested the hypothesis that the gut microbiota could induce maturation of the adult ENS through release of 5-HT and activation of 5-HT4 receptors. Colonization of germ-free mice with a microbiota from conventionally raised mice modified the neuroanatomy of the ENS and increased intestinal transit rates, which was associated with neuronal and mucosal 5-HT production and the proliferation of enteric neuronal progenitors in the adult intestine. Pharmacological modulation of the 5-HT4 receptor, as well as depletion of endogenous 5-HT, identified a mechanistic link between the gut microbiota and maturation of the adult ENS through the release of 5-HT and activation of the 5-HT4 receptor. Taken together, these findings show that the microbiota modulates the anatomy of the adult ENS in a 5-HT-dependent fashion with concomitant changes in intestinal transit.


Assuntos
Sistema Nervoso Entérico/microbiologia , Sistema Nervoso Entérico/fisiologia , Microbioma Gastrointestinal/fisiologia , Intestino Delgado/microbiologia , Serotonina/metabolismo , Animais , Sistema Nervoso Entérico/metabolismo , Feminino , Motilidade Gastrointestinal/fisiologia , Intestino Delgado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microbiota/fisiologia , Neurogênese/fisiologia , Neurônios/metabolismo , Neurônios/microbiologia , Receptores 5-HT4 de Serotonina/metabolismo
2.
Int J Mol Sci ; 22(13)2021 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-34281258

RESUMO

Infection by Proteus mirabilis causes urinary stones and catheter incrustation due to ammonia formed by urease (PMU), one of its virulence factors. Non-enzymatic properties, such as pro-inflammatory and neurotoxic activities, were previously reported for distinct ureases, including that of the gastric pathogen Helicobacter pylori. Here, PMU was assayed on isolated cells to evaluate its non-enzymatic properties. Purified PMU (nanomolar range) was tested in human (platelets, HEK293 and SH-SY5Y) cells, and in murine microglia (BV-2). PMU promoted platelet aggregation. It did not affect cellular viability and no ammonia was detected in the cultures' supernatants. PMU-treated HEK293 cells acquired a pro-inflammatory phenotype, producing reactive oxygen species (ROS) and cytokines IL-1ß and TNF-α. SH-SY5Y cells stimulated with PMU showed high levels of intracellular Ca2+ and ROS production, but unlike BV-2 cells, SH-SY5Y did not synthesize TNF-α and IL-1ß. Texas Red-labeled PMU was found in the cytoplasm and in the nucleus of all cell types. Bioinformatic analysis revealed two bipartite nuclear localization sequences in PMU. We have shown that PMU, besides urinary stone formation, can potentially contribute in other ways to pathogenesis. Our data suggest that PMU triggers pro-inflammatory effects and may affect cells beyond the renal system, indicating a possible role in extra-urinary diseases.


Assuntos
Proteus mirabilis/enzimologia , Proteus mirabilis/patogenicidade , Urease/metabolismo , Urease/toxicidade , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Células HEK293 , Humanos , Técnicas In Vitro , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/microbiologia , Modelos Moleculares , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/microbiologia , Neurotoxinas/química , Neurotoxinas/metabolismo , Neurotoxinas/toxicidade , Sinais de Localização Nuclear , Agregação Plaquetária/efeitos dos fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/toxicidade , Urease/química , Virulência/fisiologia
3.
Nano Lett ; 19(9): 5904-5914, 2019 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-31389707

RESUMO

Sensory neurons promote profound suppressive effects on neutrophils during Streptococcus pyogenes infection and contribute to the pathogenesis of necrotizing infection ("flesh-eating disease"). Thus, the development of new antibacterial agents for necrotizing infection is promising because of the clear streptococcal neuro-immune communication. Herein, based on the immune escape membrane exterior and competitive membrane functions of the glioma cell membrane, a novel nano neuro-immune blocker capsule was designed to prevent neuronal activation and improve neutrophil immune responses for necrotizing infection. These nano neuro-immune blockers could neutralize streptolysin S, suppress neuron pain conduction and calcitonin gene-related peptide release, and recruit neutrophils to the infection site, providing a strong therapeutic effect against necrotizing infection. Furthermore, nano neuro-immune blockers could serve as an effective inflammatory regulator and antibacterial agent via photothermal effects under near-infrared irradiation. In the Streptococcus pyogenes-induced necrotizing fasciitis mouse model, nano neuro-immune blockers showed significant therapeutic efficacy by ameliorating sensitivity to pain and promoting the antibacterial effect of neutrophils.


Assuntos
Antibacterianos/farmacologia , Inflamação/tratamento farmacológico , Necrose/tratamento farmacológico , Dor/tratamento farmacológico , Animais , Antibacterianos/química , Antibacterianos/efeitos da radiação , Proteínas de Bactérias/antagonistas & inibidores , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/efeitos da radiação , Inflamação/microbiologia , Luz , Camundongos , Necrose/microbiologia , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/efeitos da radiação , Neurônios/efeitos dos fármacos , Neurônios/microbiologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/microbiologia , Dor/microbiologia , Streptococcus pyogenes/efeitos dos fármacos , Streptococcus pyogenes/patogenicidade , Estreptolisinas/antagonistas & inibidores
4.
J Neurosci ; 37(48): 11758-11768, 2017 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-29089436

RESUMO

Peripheral pain signaling reflects a balance of pronociceptive and antinociceptive influences; the contribution by the gastrointestinal microbiota to this balance has received little attention. Disorders, such as inflammatory bowel disease and irritable bowel syndrome, are associated with exaggerated visceral nociceptive actions that may involve altered microbial signaling, particularly given the evidence for bacterial dysbiosis. Thus, we tested whether a community of commensal gastrointestinal bacteria derived from a healthy human donor (microbial ecosystem therapeutics; MET-1) can affect the excitability of male mouse DRG neurons. MET-1 reduced the excitability of DRG neurons by significantly increasing rheobase, decreasing responses to capsaicin (2 µm) and reducing action potential discharge from colonic afferent nerves. The increase in rheobase was accompanied by an increase in the amplitude of voltage-gated K+ currents. A mixture of bacterial protease inhibitors abrogated the effect of MET-1 effects on DRG neuron rheobase. A serine protease inhibitor but not inhibitors of cysteine proteases, acid proteases, metalloproteases, or aminopeptidases abolished the effects of MET-1. The serine protease cathepsin G recapitulated the effects of MET-1 on DRG neurons. Inhibition of protease-activated receptor-4 (PAR-4), but not PAR-2, blocked the effects of MET-1. Furthermore, Faecalibacterium prausnitzii recapitulated the effects of MET-1 on excitability of DRG neurons. We conclude that serine proteases derived from commensal bacteria can directly impact the excitability of DRG neurons, through PAR-4 activation. The ability of microbiota-neuronal interactions to modulate afferent signaling suggests that therapies that induce or correct microbial dysbiosis may impact visceral pain.SIGNIFICANCE STATEMENT Commercially available probiotics have the potential to modify visceral pain. Here we show that secretory products from gastrointestinal microbiota derived from a human donor signal to DRG neurons. Their secretory products contain serine proteases that suppress excitability via activation of protease-activated receptor-4. Moreover, from this community of commensal microbes, Faecalibacterium prausnitzii strain 16-6-I 40 fastidious anaerobe agar had the greatest effect. Our study suggests that therapies that induce or correct microbial dysbiosis may affect the excitability of primary afferent neurons, many of which are nociceptive. Furthermore, identification of the bacterial strains capable of suppressing sensory neuron excitability, and their mechanisms of action, may allow therapeutic relief for patients with gastrointestinal diseases associated with pain.


Assuntos
Gânglios Espinais/enzimologia , Microbioma Gastrointestinal/fisiologia , Granzimas/administração & dosagem , Neurônios/enzimologia , Simbiose/fisiologia , Animais , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/microbiologia , Peptídeo Hidrolases/administração & dosagem , Simbiose/efeitos dos fármacos
5.
Biochem Biophys Res Commun ; 498(4): 824-829, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29530528

RESUMO

In the mouse olfactory bulb (OB), interneurons such as granule cells and periglomerular cells are continuously replaced by adult-born neurons, which are generated in the subventricular zone (SVZ) of the brain. We have now investigated the role of commensal bacteria in regulation of such neuronal cell turnover in the adult mouse brain. Administration of mixture of antibiotics to specific pathogen-free (SPF) mice markedly attenuated the incorporation of bromodeoxyuridine (BrdU) into the SVZ cells. The treatment with antibiotics also reduced newly generated BrdU-positive neurons in the mouse OB. In addition, the incorporation of BrdU into the SVZ cells of germ-free (GF) mice was markedly reduced compared to that apparent for SPF mice. In contrast, the reduced incorporation of BrdU into the SVZ cells of GF mice was recovered by their co-housing with SPF mice, suggesting that commensal bacteria promote the incorporation of BrdU into the SVZ cells. Finally, we found that administration of ampicillin markedly attenuated the incorporation of BrdU into the SVZ cells of SPF mice. Our results thus suggest that ampicillin-sensitive commensal bacteria regulate the neurogenesis in the SVZ of adult mouse brain.


Assuntos
Fenômenos Fisiológicos Bacterianos , Ventrículos Laterais/crescimento & desenvolvimento , Ventrículos Laterais/microbiologia , Neurogênese , Bulbo Olfatório/crescimento & desenvolvimento , Bulbo Olfatório/microbiologia , Simbiose , Ampicilina/farmacologia , Animais , Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Bactérias/metabolismo , Fenômenos Fisiológicos Bacterianos/efeitos dos fármacos , Interneurônios/citologia , Interneurônios/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Neurônios/microbiologia
6.
FASEB J ; 31(7): 2785-2796, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28314768

RESUMO

Patients who recover from pneumonia subsequently have elevated rates of death after hospital discharge as a result of secondary organ damage, the causes of which are unknown. We used the bacterium Pseudomonas aeruginosa, a common cause of hospital-acquired pneumonia, as a model for investigating this phenomenon. We show that infection of pulmonary endothelial cells by P. aeruginosa induces production and release of a cytotoxic amyloid molecule with prion characteristics, including resistance to various nucleases and proteases. This cytotoxin was self-propagating, was neutralized by anti-amyloid Abs, and induced death of endothelial cells and neurons. Moreover, the cytotoxin induced edema in isolated lungs. Endothelial cells and isolated lungs were protected from cytotoxin-induced death by stimulation of signal transduction pathways that are linked to prion protein. Analysis of bronchoalveolar lavage fluid collected from human patients with P. aeruginosa pneumonia demonstrated cytotoxic activity, and lavage fluid contained amyloid molecules, including oligomeric τ and Aß. Demonstration of long-lived cytotoxic agents after Pseudomonas infection may establish a molecular link to the high rates of death as a result of end-organ damage in the months after recovery from pneumonia, and modulation of signal transduction pathways that have been linked to prion protein may provide a mechanism for intervention.-Balczon, R., Morrow, K. A., Zhou, C., Edmonds, B., Alexeyev, M., Pittet, J.-F., Wagener, B. M., Moser, S. A., Leavesley, S., Zha, X., Frank, D. W., Stevens, T. Pseudomonas aeruginosa infection liberates transmissible, cytotoxic prion amyloids.


Assuntos
Citotoxinas/metabolismo , Proteínas Priônicas/toxicidade , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa , Animais , Edema , Células Endoteliais/microbiologia , Humanos , Camundongos , Neurônios/microbiologia , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia , Proteínas Priônicas/metabolismo , Infecções por Pseudomonas/patologia , Ratos
7.
Brain Behav Immun ; 67: 218-229, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28890156

RESUMO

The mammalian fetus develops in a largely sterile environment, and direct exposure to a complex microbiota does not occur until birth. We took advantage of this to examine the effect of the microbiota on brain development during the first few days of life. The expression of anti- and pro-inflammatory cytokines, developmental cell death, and microglial colonization in the brain were compared between newborn conventionally colonized mice and mice born in sterile, germ-free (GF) conditions. Expression of the pro-inflammatory cytokines interleukin 1ß and tumor necrosis factor α was markedly suppressed in GF newborns. GF mice also had altered cell death, with some regions exhibiting higher rates (paraventricular nucleus of the hypothalamus and the CA1 oriens layer of the hippocampus) and other regions exhibiting no change or lower rates (arcuate nucleus of the hypothalamus) of cell death. Microglial labeling was elevated in GF mice, due to an increase in both microglial cell size and number. The changes in cytokine expression, cell death and microglial labeling were evident on the day of birth, but were absent on embryonic day 18.5, approximately one-half day prior to expected delivery. Taken together, our results suggest that direct exposure to the microbiota at birth influences key neurodevelopmental events and does so within hours. These findings may help to explain some of the behavioral and neurochemical alterations previously seen in adult GF mice.


Assuntos
Encéfalo/crescimento & desenvolvimento , Morte Celular , Encefalite/microbiologia , Microbiota , Microglia/fisiologia , Neurônios/fisiologia , Animais , Encéfalo/microbiologia , Encefalite/metabolismo , Feminino , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Microglia/microbiologia , Neurônios/microbiologia , Gravidez
8.
Infect Immun ; 85(1)2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27795365

RESUMO

Botulinum neurotoxins (BoNTs) are the most toxic proteins for humans but also are common therapies for neurological diseases. BoNTs are dichain toxins, comprising an N-terminal catalytic domain (LC) disulfide bond linked to a C-terminal heavy chain (HC) which includes a translocation domain (HN) and a receptor binding domain (HC). Recently, the BoNT serotype A (BoNT/A) subtypes A1 and A2 were reported to possess similar potencies but different rates of cellular intoxication and pathology in a mouse model of botulism. The current study measured HCA1 and HCA2 entry into rat primary neurons and cultured Neuro2A cells. We found that there were two sequential steps during the association of BoNT/A with neurons. The initial step was ganglioside dependent, while the subsequent step involved association with synaptic vesicles. HCA1 and HCA2 entered the same population of synaptic vesicles and entered cells at similar rates. The primary difference was that HCA2 had a higher degree of receptor occupancy for cells and neurons than HcA1. Thus, HCA2 and HCA1 share receptors and entry pathway but differ in their affinity for receptor. The initial interaction of HCA1 and HCA2 with neurons may contribute to the unique pathologies of BoNT/A1 and BoNT/A2 in mouse models.


Assuntos
Toxinas Botulínicas Tipo A/metabolismo , Botulismo/metabolismo , Botulismo/microbiologia , Neurônios/metabolismo , Neurônios/microbiologia , Animais , Células Cultivadas , Clostridium botulinum/patogenicidade , Gangliosídeos/metabolismo , Camundongos , Ligação Proteica/fisiologia , Ratos , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/microbiologia
9.
Neurobiol Dis ; 108: 249-260, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28888971

RESUMO

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease and the main cause of motor neuron pathology. The etiology of the disease remains unknown, and no effective therapy exists to halt the disease or improve the quality of life. Here, we provide compelling evidence for the existence of fungal infection in ALS. Immunohistochemistry analysis using a battery of antifungal antibodies revealed fungal structures such as yeast and hyphae in the motor cortex, the medulla and the spinal cord, in eleven patients with ALS. Some fungal structures were localized intracellularly and even intranuclearly, indicating that this infection is not the result of post-mortem colonization. By contrast, this burden of fungal infection cannot be observed in several CNS areas of control subjects. PCR analysis and next generation sequencing of DNA extracted from frozen neural tissue identified a variety of fungal genera including Candida, Malassezia, Fusarium, Botrytis, Trichoderma and Cryptococcus. Overall, our present observations provide strong evidence for mixed fungal infections in ALS patients. The exact mixed infection varies from patient to patient consistent with the different evolution and severity of symptoms in each ALS patient. These novel findings provide a logical explanation for the neuropathological observations of this disease, such as neuroinflammation and elevated chitinase levels, and could help to implement appropriate therapies.


Assuntos
Esclerose Lateral Amiotrófica/complicações , Esclerose Lateral Amiotrófica/patologia , Infecções Fúngicas do Sistema Nervoso Central/complicações , Infecções Fúngicas do Sistema Nervoso Central/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Esclerose Lateral Amiotrófica/microbiologia , Feminino , Genômica , Humanos , Imuno-Histoquímica , Masculino , Bulbo/microbiologia , Bulbo/patologia , Metagenoma , Pessoa de Meia-Idade , Córtex Motor/microbiologia , Córtex Motor/patologia , Neurônios/microbiologia , Neurônios/patologia , Reação em Cadeia da Polimerase , Análise de Sequência , Medula Espinal/microbiologia , Medula Espinal/patologia
10.
J Cell Sci ; 127(Pt 15): 3294-308, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24928902

RESUMO

Proteolytic cleavage of synaptosomal-associated protein 25 by the light chain of botulinum neurotoxin type A (LCA) results in a blockade of neurotransmitter release that persists for several months in motor neurons. The L428A/L429A mutation in LCA is known to significantly shorten both the proteolytic and neuroparalytic effects of the neurotoxin in mice. To elucidate the cellular mechanism for LCA longevity, we studied the effects of L428A/L429A mutation on the interactome, localization and stability of LCA expressed in cultured neuronal cells. Mass spectrometry analysis of the LCA interactome showed that the mutation prevented the interaction of LCA with septins. The wild-type LCA was concentrated in plasma-membrane-associated clusters, colocalizing with septins-2 and septin-7, which accumulated in these clusters only in the presence of LCA. The L428A/L429A mutation decreased co-clustering of LCA and septins and accelerated proteasomal and non-proteasomal degradation of LCA. Similarly, the impairment of septin oligomerization by forchlorfenuron or silencing of septin-2 prevented LCA interaction and clustering with septins and increased LCA degradation. Therefore, the dileucine-mediated LCA-septin co-clustering is crucial for the long-lasting stabilization of LCA-related proteolytic and presumably neuroparalytic activity.


Assuntos
Toxinas Botulínicas Tipo A/metabolismo , Membrana Celular/metabolismo , Neurônios/fisiologia , Síndromes Neurotóxicas/metabolismo , Septinas/metabolismo , Animais , Toxinas Botulínicas Tipo A/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Humanos , Camundongos , Mutação/genética , Neurônios/microbiologia , Síndromes Neurotóxicas/microbiologia , Compostos de Fenilureia/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Multimerização Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Piridinas/farmacologia , RNA Interferente Pequeno/genética , Septinas/genética
11.
Nat Rev Neurosci ; 12(6): 345-57, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21587289

RESUMO

The nervous system is protected by barriers that restrict the invasion of pathogens. Nevertheless, mechanisms have evolved by which microbes can pass these barriers, enter and exit neurons and target various regions of the nervous system. In the brain, immune responses to pathogens are generally not robust, so microbes can hide and survive or, conversely, cause severe uncontrolled infections. Depending on their sites of entry and the regions that they target, microbes can cause diverse nervous system dysfunctions and even influence host behaviour to their own advantage. This Review discusses routes by which microbes can reach the nervous system and cause persistent or life-threatening infections.


Assuntos
Barreira Hematoencefálica/microbiologia , Encéfalo/microbiologia , Neurônios/microbiologia , Animais
12.
PLoS Genet ; 9(3): e1003324, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23505381

RESUMO

In Caenorhabditis elegans the Toll-interleukin receptor domain adaptor protein TIR-1 via a conserved mitogen-activated protein kinase (MAPK) signaling cascade induces innate immunity and upregulates serotonin (5-HT) biosynthesis gene tph-1 in a pair of ADF chemosensory neurons in response to infection. Here, we identify transcription factors downstream of the TIR-1 signaling pathway. We show that common transcription factors control the innate immunity and 5-HT biosynthesis. We demonstrate that a cysteine to tyrosine substitution in an ARM motif of the HEAT/Arm repeat region of the TIR-1 protein confers TIR-1 hyperactivation, leading to constitutive tph-1 upregulation in the ADF neurons, increased expression of intestinal antimicrobial genes, and enhanced resistance to killing by the human opportunistic pathogen Pseudomonas aeruginosa PA14. A forward genetic screen for suppressors of the hyperactive TIR-1 led to the identification of DAF-19, an ortholog of regulatory factor X (RFX) transcription factors that are required for human adaptive immunity. We show that DAF-19 concerts with ATF-7, a member of the activating transcription factor (ATF)/cAMP response element-binding B (CREB) family of transcription factors, to regulate tph-1 and antimicrobial genes, reminiscent of RFX-CREB interaction in human immune cells. daf-19 mutants display heightened susceptibility to killing by PA14. Remarkably, whereas the TIR-1-MAPK-DAF-19/ATF-7 pathway in the intestinal immunity is regulated by DKF-2/protein kinase D, we found that the regulation of tph-1 expression is independent of DKF-2 but requires UNC-43/Ca(2+)/calmodulin-dependent protein kinase (CaMK) II. Our results suggest that pathogenic cues trigger a common core-signaling pathway via tissue-specific mechanisms and demonstrate a novel role for RFX factors in neuronal and innate immune responses to infection.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Imunidade Inata , Pseudomonas aeruginosa , Serotonina , Fatores de Transcrição , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/imunologia , Caenorhabditis elegans/microbiologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/imunologia , Proteínas de Caenorhabditis elegans/metabolismo , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Humanos , Intestinos/imunologia , Intestinos/microbiologia , Neurônios/citologia , Neurônios/metabolismo , Neurônios/microbiologia , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidade , Receptores Acoplados a Proteínas G , Serotonina/biossíntese , Serotonina/genética , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Triptofano Hidroxilase/metabolismo
13.
Immunol Cell Biol ; 93(9): 789-95, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25801479

RESUMO

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.


Assuntos
Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Adipócitos/imunologia , Adipócitos/microbiologia , Células Endoteliais/imunologia , Células Endoteliais/microbiologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Fibroblastos/imunologia , Fibroblastos/microbiologia , Humanos , Modelos Imunológicos , Mycobacterium tuberculosis/fisiologia , Neuroglia/imunologia , Neuroglia/microbiologia , Neurônios/imunologia , Neurônios/microbiologia , Tuberculose/microbiologia
14.
J Neuroinflammation ; 12: 125, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-26112704

RESUMO

BACKGROUND: Tuberculosis (TB) affects one third of the global population, and TB of the central nervous system (CNS-TB) is the most severe form of tuberculosis which often associates with high mortality. The pro-inflammatory cytokine tumour necrosis factor (TNF) plays a critical role in the initial and long-term host immune protection against Mycobacterium tuberculosis (M. tuberculosis) which involves the activation of innate immune cells and structure maintenance of granulomas. However, the contribution of TNF, in particular neuron-derived TNF, in the control of cerebral M. tuberculosis infection and its protective immune responses in the CNS were not clear. METHODS: We generated neuron-specific TNF-deficient (NsTNF(-/-)) mice and compared outcomes of disease against TNF(f/f) control and global TNF(-/-) mice. Mycobacterial burden in brains, lungs and spleens were compared, and cerebral pathology and cellular contributions analysed by microscopy and flow cytometry after M. tuberculosis infection. Activation of innate immune cells was measured by flow cytometry and cell function assessed by cytokine and chemokine quantification using enzyme-linked immunosorbent assay (ELISA). RESULTS: Intracerebral M. tuberculosis infection of TNF(-/-) mice rendered animals highly susceptible, accompanied by uncontrolled bacilli replication and eventual mortality. In contrast, NsTNF(-/-) mice were resistant to infection and presented with a phenotype similar to that in TNF(f/f) control mice. Impaired immunity in TNF(-/-) mice was associated with altered cytokine and chemokine synthesis in the brain and characterised by a reduced number of activated innate immune cells. Brain pathology reflected enhanced inflammation dominated by neutrophil influx. CONCLUSION: Our data show that neuron-derived TNF has a limited role in immune responses, but overall TNF production is necessary for protective immunity against CNS-TB.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Imunidade Inata/fisiologia , Mycobacterium tuberculosis/fisiologia , Neurônios/microbiologia , Neurônios/patologia , Tuberculose do Sistema Nervoso Central/imunologia , Fator de Necrose Tumoral alfa/fisiologia , Replicação Viral/fisiologia , Animais , Encéfalo/metabolismo , Encéfalo/microbiologia , Encéfalo/patologia , Proliferação de Células/fisiologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Dendríticas/microbiologia , Células Dendríticas/patologia , Modelos Animais de Doenças , Resistência à Doença/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Camundongos Knockout , Microglia/microbiologia , Microglia/patologia , Tuberculose do Sistema Nervoso Central/patologia , Tuberculose do Sistema Nervoso Central/fisiopatologia , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética
15.
Brain Behav Immun ; 50: 166-177, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26144888

RESUMO

There are extensive bidirectional interactions between the gut microbiota and the central nervous system (CNS), and studies demonstrate that stressor exposure significantly alters gut microbiota community structure. We tested whether oligosaccharides naturally found in high levels in human milk, which have been reported to impact brain development and enhance the growth of beneficial commensal microbes, would prevent stressor-induced alterations in gut microbial community composition and attenuate stressor-induced anxiety-like behavior. Mice were fed standard laboratory diet, or laboratory diet containing the human milk oligosaccharides 3'Sialyllactose (3'SL) or 6'Sialyllactose (6'SL) for 2 weeks prior to being exposed to either a social disruption stressor or a non-stressed control condition. Stressor exposure significantly changed the structure of the colonic mucosa-associated microbiota in control mice, as indicated by changes in beta diversity. The stressor resulted in anxiety-like behavior in both the light/dark preference and open field tests in control mice. This effect was associated with a reduction in immature neurons in the dentate gyrus as indicated by doublecortin (DCX) immunostaining. These effects were not evident in mice fed milk oligosaccharides; stressor exposure did not significantly change microbial community structure in mice fed 3'SL or 6'SL. In addition, 3'SL and 6'SL helped maintain normal behavior on tests of anxiety-like behavior and normal numbers of DCX+ immature neurons. These studies indicate that milk oligosaccharides support normal microbial communities and behavioral responses during stressor exposure, potentially through effects on the gut microbiota-brain axis.


Assuntos
Ansiedade/microbiologia , Encéfalo/microbiologia , Microbioma Gastrointestinal/fisiologia , Lactose/análogos & derivados , Oligossacarídeos/administração & dosagem , Estresse Psicológico/microbiologia , Animais , Índice de Massa Corporal , Proliferação de Células , Corticosterona/sangue , Proteína Duplacortina , Interleucina-6/sangue , Lactose/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/microbiologia , Baço/microbiologia
16.
Genesis ; 52(9): 827-832, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25112520

RESUMO

LHX9 is a LIM-homeodomain transcription factor essential for the development of gonads, spinal cord interneurons, and thalamic neurons to name a few. We recently reported the expression of LHX9 in retinal amacrine cells during development. In this study, we generated an Lhx9-GFPCreER(T) (2) (GCE) knock-in mouse line by knocking-in a GCE cassette at the Lhx9 locus, thus inactivating endogenous Lhx9. Lhx9(GCE) (/+) mice were viable, fertile, and displayed no overt phenotypical characteristics. Lhx9(GCE) (/) (GCE) mice were all phenotypically female, smaller in size, viable, but infertile. The specificity and efficacy of the Lhx9-GCE mouse line was verified by crossing it to a Rosa26-tdTomato reporter mouse line, which reveals the Cre recombinase activities in retinal amacrine cells, developing limbs, testis, hippocampal neurons, thalamic neurons, and cerebellar neurons. Taken together, the Lhx9-GCE mouse line could serve as a beneficial tool for lineage tracing and gene manipulation experiments. genesis


Assuntos
Proteínas com Homeodomínio LIM/genética , Fatores de Transcrição/genética , Células Amácrinas/citologia , Células Amácrinas/metabolismo , Animais , Extremidades/embriologia , Extremidades/crescimento & desenvolvimento , Feminino , Técnicas de Introdução de Genes , Loci Gênicos , Hipocampo/embriologia , Hipocampo/crescimento & desenvolvimento , Integrases/genética , Integrases/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Masculino , Camundongos , Neurônios/metabolismo , Neurônios/microbiologia , Retina/citologia , Retina/embriologia , Retina/crescimento & desenvolvimento , Testículo/embriologia , Testículo/crescimento & desenvolvimento , Fatores de Transcrição/metabolismo
17.
Infect Immun ; 82(5): 1880-90, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24566619

RESUMO

Mycobacterium tuberculosis infection of the central nervous system is thought to be initiated once the bacilli have breached the blood brain barrier and are phagocytosed, primarily by microglial cells. In this study, the interactions of M. tuberculosis with neurons in vitro and in vivo were investigated. The data obtained demonstrate that neurons can act as host cells for M. tuberculosis. M. tuberculosis bacilli were internalized by murine neuronal cultured cells in a time-dependent manner after exposure, with superior uptake by HT22 cells compared to Neuro-2a cells (17.7% versus 9.8%). Internalization of M. tuberculosis bacilli by human SK-N-SH cultured neurons suggested the clinical relevance of the findings. Moreover, primary murine hippocampus-derived neuronal cultures could similarly internalize M. tuberculosis. Internalized M. tuberculosis bacilli represented a productive infection with retention of bacterial viability and replicative potential, increasing 2- to 4-fold within 48 h. M. tuberculosis bacillus infection of neurons was confirmed in vivo in the brains of C57BL/6 mice after intracerebral challenge. This study, therefore, demonstrates neurons as potential new target cells for M. tuberculosis within the central nervous system.


Assuntos
Mycobacterium tuberculosis/fisiologia , Neurônios/microbiologia , Tuberculose do Sistema Nervoso Central/microbiologia , Animais , Linhagem Celular , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Tuberculose do Sistema Nervoso Central/imunologia
18.
Curr Top Microbiol Immunol ; 364: 159-77, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23239353

RESUMO

Synaptic vesicles (SV) are key organelles of neuronal communication. SV are responsible for the storage of neurotransmitters, which are released by Ca(2+)-dependent exocytosis. After release and interaction with postsynaptic receptors, transmitters rapidly diffuse out of the synaptic cleft and are sequestered by plasma membrane transporters (in some cases following enzymatic conversion). SVs undergo endocytosis and are refilled by specific vesicular transmitter transporters different in the various neuronal subtypes. Besides these differences, SVs in general are equipped with a remarkable common set of proteins. Botulinum neurotoxins (BoNTs) inhibit neurotransmitter release from almost all types of neurons by cleaving proteins required for membrane fusion localized either to SVs (synaptobrevin) or to the plasma membrane (SNAP-25 and syntaxin) depending on the BoNT serotype. To enter the neuronal cytoplasm, BoNTs specifically interact with the luminal domain of SV proteins (synaptotagmin or SV2, depending on serotype) transiently exposed during exocytotic membrane fusion and occurring in almost every neuron. Thus, the highly specific interaction with luminal domains of SV proteins commonly expressed on all SV types is one reason why BoNTs exhibit such a high neuronal specificity but attack almost every neuron type.


Assuntos
Toxinas Botulínicas/metabolismo , Neurotoxinas/metabolismo , Vesículas Sinápticas/metabolismo , Animais , Toxinas Botulínicas/toxicidade , Botulismo/microbiologia , Botulismo/fisiopatologia , Membrana Celular/metabolismo , Clostridium botulinum/patogenicidade , Exocitose , Fusão de Membrana , Glicoproteínas de Membrana/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/microbiologia , Neurotoxinas/toxicidade , Transporte Proteico , Proteólise , Transmissão Sináptica , Proteína 25 Associada a Sinaptossoma/metabolismo , Sinaptotagminas/metabolismo
19.
Cell Microbiol ; 15(5): 742-58, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23152983

RESUMO

Headache, muscle aches and chest pain of mild to medium intensity are among the most common clinical symptoms in moderate Staphylococcus aureus infections, with severe infections usually associated with worsening pain symptoms. These nociceptive responses of the body raise the question of how bacterial infection impinges on the nervous system. Does S. aureus, or its released virulence factors, act directly on neurones? To address this issue, we evaluated the potential effects on neurones of certain bi-component leukotoxins, which are virulent factors released by the bacterium. The activity of four different leukotoxins was verified by measuring the release of glutamate from rat cerebellar granular neurones. The bi-component γ-haemolysin HlgC/HlgB was the most potent leukotoxin, initiating transient rises in intracellular Ca(2+) concentration in cerebellar neurones and in primary sensory neurones from dorsal root ganglia, as probed with the Fura-2 Ca(2+) indicator dye. Using pharmacological antagonists of receptors and Ca(2+) channels, the variations in intracellular Ca(2+) concentration were found independent of the activation of voltage-operated Ca(2+) channels or glutamate receptors. Drugs targeting Sarco-Endoplasmic Reticulum Ca(2+)-ATPase (SERCA) or H(+)-ATPase and antagonists of the store-operated Ca(2+) entry complex blunted, or significantly reduced, the leukotoxin-induced elevation in intracellular Ca(2+). Moreover, activation of the ADP-ribosyl cyclase CD38 was also required to initiate the release of Ca(2+) from acidic stores. These findings suggest that, prior to forming a pore at the plasma membrane, leukotoxin HlgC/HlgB triggers a multistep process which initiates the release of Ca(2+) from lysosomes, modifies the steady-state level of reticular Ca(2+) stores and finally activates the Store-Operated Calcium Entry complex.


Assuntos
Proteínas de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Cálcio/metabolismo , Proteínas Hemolisinas/farmacologia , Neurônios/metabolismo , Staphylococcus aureus/patogenicidade , Animais , Cafeína/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/microbiologia , Gânglios/metabolismo , Gânglios/microbiologia , Gânglios Espinais/metabolismo , Ácido Glutâmico/metabolismo , Humanos , Neurônios/efeitos dos fármacos , Neurônios/microbiologia , ATPases Translocadoras de Prótons/metabolismo , Ratos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Transdução de Sinais/efeitos dos fármacos , Staphylococcus aureus/genética
20.
Biochem J ; 445(2): 205-12, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22519640

RESUMO

Caenopores are antimicrobial and pore-forming polypeptides in Caenorhabditis elegans belonging to the saposin-like protein superfamily and are considered important elements of the nematode's intestinal immune system. In the present study, we demonstrate that, unlike the other members of the multifarious gene family (spps) coding for caenopores, spp-12 is expressed exclusively in two pharyngeal neurons. Recombinantly expressed SPP-12 binds to phospholipid membranes and forms pores in a pH-dependent manner characteristic of caenopores. Moreover, SPP-12 kills viable Gram-positive bacteria, yeast cells and amoebae by permeabilizing their membranes, suggesting a wide-target cell spectrum. A spp-12 knockout mutant is more susceptible to pathogenic Bacillus thuringiensis than wild-type worms and is tolerant to non-pathogenic bacteria. By contrast, SPP-1, a caenopore, whose gene is expressed only in the intestine and reported to be regulated by the same pathway as spp-12, is apparently non-protective against pathogenic B. thuringiensis, although it also does display antimicrobial activity. The transcription of spp-1 is down-regulated in wild-type worms in the presence of pathogenic B. thuringiensis and a spp-1 knockout mutant is hyposusceptible to this bacterium. This implies that SPP-12, but not SPP-1, contributes to resistance against B. thuringiensis, a natural pathogen of the nematode.


Assuntos
Bacillus thuringiensis/patogenicidade , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/microbiologia , Neurônios/metabolismo , Faringe/metabolismo , Envelhecimento/fisiologia , Animais , Animais Geneticamente Modificados , Anti-Infecciosos/farmacologia , Proteínas de Caenorhabditis elegans/genética , Interações Hospedeiro-Patógeno , Técnicas Imunoenzimáticas , Neurônios/microbiologia , Peptídeos/farmacologia , Faringe/microbiologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA