Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Chem Senses ; 44(8): 615-630, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31403159

ABSTRACT

Sensory systems such as the olfactory system detect chemical stimuli and thereby determine the relationships between the animal and its surroundings. Olfaction is one of the most conserved and ancient sensory systems in vertebrates. The vertebrate olfactory epithelium is colonized by complex microbial communities, but microbial contribution to host olfactory gene expression remains unknown. In this study, we show that colonization of germ-free zebrafish and mice with microbiota leads to widespread transcriptional responses in olfactory organs as measured in bulk tissue transcriptomics and RT-qPCR. Germ-free zebrafish olfactory epithelium showed defects in pseudostratification; however, the size of the olfactory pit and the length of the cilia were not different from that of colonized zebrafish. One of the mechanisms by which microbiota control host transcriptional programs is by differential expression and activity of specific transcription factors (TFs). REST (RE1 silencing transcription factor, also called NRSF) is a zinc finger TF that binds to the conserved motif repressor element 1 found in the promoter regions of many neuronal genes with functions in neuronal development and differentiation. Colonized zebrafish and mice showed increased nasal expression of REST, and genes with reduced expression in colonized animals were strongly enriched in REST-binding motifs. Nasal commensal bacteria promoted in vitro differentiation of Odora cells by regulating the kinetics of REST expression. REST knockdown resulted in decreased Odora cell differentiation in vitro. Our results identify a conserved mechanism by which microbiota regulate vertebrate olfactory transcriptional programs and reveal a new role for REST in sensory organs.


Subject(s)
Microbiota/physiology , Nerve Tissue Proteins/genetics , Olfactory Mucosa/metabolism , Olfactory Receptor Neurons/metabolism , Repressor Proteins/genetics , Smell/genetics , Animals , Cell Line , Conserved Sequence , Gene Expression Profiling , Gene Expression Regulation , Germ-Free Life , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Olfactory Mucosa/cytology , Olfactory Mucosa/microbiology , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/microbiology , Promoter Regions, Genetic , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Repressor Proteins/metabolism , Symbiosis/physiology , Zebrafish
2.
Infect Immun ; 84(7): 1941-1956, 2016 07.
Article in English | MEDLINE | ID: mdl-27091931

ABSTRACT

Melioidosis, caused by the bacterium Burkholderia pseudomallei, is an often severe infection that regularly involves respiratory disease following inhalation exposure. Intranasal (i.n.) inoculation of mice represents an experimental approach used to study the contributions of bacterial capsular polysaccharide I (CPS I) to virulence during acute disease. We used aerosol delivery of B. pseudomallei to establish respiratory infection in mice and studied CPS I in the context of innate immune responses. CPS I improved B. pseudomallei survival in vivo and triggered multiple cytokine responses, neutrophil infiltration, and acute inflammatory histopathology in the spleen, liver, nasal-associated lymphoid tissue, and olfactory mucosa (OM). To further explore the role of the OM response to B. pseudomallei infection, we infected human olfactory ensheathing cells (OECs) in vitro and measured bacterial invasion and the cytokine responses induced following infection. Human OECs killed >90% of the B. pseudomallei in a CPS I-independent manner and exhibited an antibacterial cytokine response comprising granulocyte colony-stimulating factor, tumor necrosis factor alpha, and several regulatory cytokines. In-depth genome-wide transcriptomic profiling of the OEC response by RNA-Seq revealed a network of signaling pathways activated in OECs following infection involving a novel group of 378 genes that encode biological pathways controlling cellular movement, inflammation, immunological disease, and molecular transport. This represents the first antimicrobial program to be described in human OECs and establishes the extensive transcriptional defense network accessible in these cells. Collectively, these findings show a role for CPS I in B. pseudomallei survival in vivo following inhalation infection and the antibacterial signaling network that exists in human OM and OECs.


Subject(s)
Bacterial Capsules/immunology , Burkholderia pseudomallei/immunology , Host-Pathogen Interactions/immunology , Melioidosis/immunology , Melioidosis/microbiology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Animals , Bacterial Capsules/genetics , Bacterial Load , Burkholderia pseudomallei/genetics , Cells, Cultured , Computational Biology/methods , Cytokines/metabolism , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Immunity, Innate , Melioidosis/genetics , Melioidosis/metabolism , Mice , Mutation , Neutrophil Infiltration , Olfactory Receptor Neurons/immunology , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/microbiology , Respiratory Tract Infections/genetics , Respiratory Tract Infections/metabolism , Signal Transduction , Virulence , Virulence Factors
3.
J Gen Virol ; 75 ( Pt 9): 2319-27, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8077930

ABSTRACT

One-week-old pigs were infected intranasally with the Ka strain of Aujeszky's disease virus (ADV) or with mutants that were lacking the non-essential envelope glycoproteins gI, gp63 or gIII. The invasion and spread of these strains in the olfactory nervous pathway were examined by assessing virus levels and by localizing viral antigens in the olfactory mucosa representing the first neuronal level, in the olfactory bulb representing the second neuronal level and in the lateral olfactory gyrus, the rostral perforated substance and the piriform lobe, all representing the third neuronal level. The Ka parental strain invaded and spread up to the third neuronal level. The extent of invasion and spread of the gIII- mutant were similar to those of the parental strain. The gp63- mutant replicated normally in the olfactory mucosa, but its spread to all the other levels was limited as compared with that of the parental strain. The gI- mutant showed a defect in infection at all neuronal levels. These results indicate that, of the non-essential envelope glycoproteins, gI plays the major role in neural invasion and spread of ADV in its natural host. The pattern of invasion and spread of these mutants in the olfactory pathway of pigs was similar to that previously observed in the trigeminal pathway. The type of nervous pathway therefore appears not to influence the neuropathogenesis of ADV or mutants deleted in non-essential envelope glycoproteins in the pig.


Subject(s)
Gene Deletion , Herpesvirus 1, Suid/physiology , Olfactory Bulb/microbiology , Olfactory Pathways/microbiology , Viral Envelope Proteins/metabolism , Virus Replication , Animals , Animals, Newborn , Herpesvirus 1, Suid/genetics , Immunoenzyme Techniques , Neurons/microbiology , Olfactory Receptor Neurons/microbiology , Species Specificity , Swine , Viral Envelope Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...