Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Microbiologyopen ; 8(10): e858, 2019 10.
Article in English | MEDLINE | ID: mdl-31197972

ABSTRACT

The binary association between the squid, Euprymna scolopes, and its symbiont, Vibrio fischeri, serves as a model system to study interactions between beneficial bacteria and the innate immune system. Previous research demonstrated that binding of the squid's immune cells, hemocytes, to V. fischeri is altered if the symbiont is removed from the light organ, suggesting that host colonization alters hemocyte recognition of V. fischeri. To investigate the influence of symbiosis on immune maturation during development, we characterized hemocyte binding and phagocytosis of V. fischeri and nonsymbiotic Vibrio harveyi from symbiotic (sym) and aposymbiotic (apo) juveniles, and wild-caught and laboratory-raised sym and apo adults. Our results demonstrate that while light organ colonization by V. fischeri did not alter juvenile hemocyte response, these cells bound a similar number of V. fischeri and V. harveyi yet phagocytosed only V. harveyi. Our results also indicate that long-term colonization altered the adult hemocyte response to V. fischeri but not V. harveyi. All hemocytes from adult squid, regardless of apo or sym state, both bound and phagocytosed a similar number of V. harveyi while hemocytes from both wild-caught and sym-raised adults bound significantly fewer V. fischeri, although more V. fischeri were phagocytosed by hemocytes from wild-caught animals. In contrast, hemocytes from apo-raised squid bound similar numbers of both V. fischeri and V. harveyi, although more V. harveyi cells were engulfed, suggesting that blood cells from apo-raised adults behaved similarly to juvenile hosts. Taken together, these data suggest that persistent colonization by the light organ symbiont is required for hemocytes to differentially bind and phagocytose V. fischeri. The cellular immune system of E. scolopes likely possesses multiple mechanisms at different developmental stages to promote a specific and life-long interaction with the symbiont.


Subject(s)
Aliivibrio fischeri/growth & development , Aliivibrio fischeri/immunology , Decapodiformes/immunology , Decapodiformes/microbiology , Hemocytes/immunology , Immunity, Innate , Symbiosis , Animals , Bacterial Adhesion , Cell Differentiation , Phagocytosis , Vibrio/growth & development , Vibrio/immunology
2.
mBio ; 8(2)2017 04 04.
Article in English | MEDLINE | ID: mdl-28377525

ABSTRACT

We characterized bactericidal permeability-increasing proteins (BPIs) of the squid Euprymna scolopes, EsBPI2 and EsBPI4. They have molecular characteristics typical of other animal BPIs, are closely related to one another, and nest phylogenetically among invertebrate BPIs. Purified EsBPIs had antimicrobial activity against the squid's symbiont, Vibrio fischeri, which colonizes light organ crypt epithelia. Activity of both proteins was abrogated by heat treatment and coincubation with specific antibodies. Pretreatment under acidic conditions similar to those during symbiosis initiation rendered V. fischeri more resistant to the antimicrobial activity of the proteins. Immunocytochemistry localized EsBPIs to the symbiotic organ and other epithelial surfaces interacting with ambient seawater. The proteins differed in intracellular distribution. Further, whereas EsBPI4 was restricted to epithelia, EsBPI2 also occurred in blood and in a transient juvenile organ that mediates hatching. The data provide evidence that these BPIs play different defensive roles early in the life of E. scolopes, modulating interactions with the symbiont.IMPORTANCE This study describes new functions for bactericidal permeability-increasing proteins (BPIs), members of the lipopolysaccharide-binding protein (LBP)/BPI protein family. The data provide evidence that these proteins play a dual role in the modulation of symbiotic bacteria. In the squid-vibrio model, these proteins both control the symbiont populations in the light organ tissues where symbiont cells occur in dense monoculture and, concomitantly, inhibit the symbiont from colonizing other epithelial surfaces of the animal.


Subject(s)
Aliivibrio fischeri/growth & development , Aliivibrio fischeri/immunology , Anti-Infective Agents/metabolism , Antimicrobial Cationic Peptides/metabolism , Blood Proteins/metabolism , Decapodiformes/immunology , Decapodiformes/microbiology , Symbiosis , Animals
3.
Microb Cell Fact ; 14: 37, 2015 Mar 18.
Article in English | MEDLINE | ID: mdl-25888727

ABSTRACT

BACKGROUND: Delivery of antigens by live bacterial carriers can elicit effective humoral and cellular responses and may be an attractive strategy for live bacterial vaccine production through introduction of a vector that expresses an exogenous protective antigen. To overcome the instability and metabolic burden associated with plasmid introduction, alternative strategies, such as the use of in vivo-inducible promoters, have been proposed. However, screening an ideal in vivo-activated promoter with high efficiency and low leak expression in a particular strain poses great challenges to many researchers. RESULTS: In this work, we constructed an in vivo antigen-expressing vector suitable for Edwardsiella tarda, an enteric Gram-negative invasive intracellular pathogen of both animals and humans. By combining quorum sensing genes from Vibrio fischeri with iron uptake regulons, a synthetic binary regulation system (ironQS) for E. tarda was designed. In vitro expression assay demonstrated that the ironQS system is only initiated in the absence of Fe2+ in the medium when the cell density reaches its threshold. The ironQS system was further confirmed in vivo to present an in vivo-triggered and cell density-dependent expression pattern in larvae and adult zebrafish. A recombinant E. tarda vector vaccine candidate WED(ironQS-G) was established by introducing gapA34, which encodes the protective antigen glyceraldehyde-3-phosphate dehydrogenase (GAPDH) from the fish pathogen Aeromonas hydrophila LSA34 into ironQS system, and the immune protection afforded by this vaccine was assessed in turbot (Scophtalmus maximus). Most of the vaccinated fish survived under the challenge with A. hydrophila LSA34 (RPS=67.0%) or E. tarda EIB202 (RPS=72.3%). CONCLUSIONS: Quorum sensing system has been extensively used in various gene structures in synthetic biology as a well-functioning and population-dependent gene circuit. In this work, the in vivo expression system, ironQS, maintained the high expression efficiency of the quorum sensing circuit and achieved excellent expression regulation of the Fur box. The ironQS system has great potential in applications requiring in vivo protein expression, such as vector vaccines. Considering its high compatibility, ironQS system could function as a universal expression platform for a variety of bacterial hosts.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Edwardsiella tarda/immunology , Glyceraldehyde-3-Phosphate Dehydrogenases/immunology , Quorum Sensing/immunology , Aeromonas hydrophila/genetics , Aeromonas hydrophila/immunology , Aliivibrio fischeri/genetics , Aliivibrio fischeri/immunology , Animals , Antigens, Bacterial/genetics , Bacterial Vaccines/genetics , Edwardsiella tarda/genetics , Fish Diseases/immunology , Fish Diseases/mortality , Fish Diseases/parasitology , Flatfishes/immunology , Flatfishes/parasitology , Gene Expression/drug effects , Gene Expression/immunology , Genetic Vectors/genetics , Genetic Vectors/immunology , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Iron/pharmacology , Larva/immunology , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Quorum Sensing/genetics , Reproducibility of Results , Survival Rate , Vaccination/methods , Zebrafish/immunology
4.
PLoS One ; 10(3): e0119949, 2015.
Article in English | MEDLINE | ID: mdl-25775132

ABSTRACT

In the mutualistic relationship between the squid Euprymna tasmanica and the bioluminescent bacterium Vibrio fischeri, several host factors, including immune-related proteins, are known to interact and respond specifically and exclusively to the presence of the symbiont. In squid and octopus, the white body is considered to be an immune organ mainly due to the fact that blood cells, or hemocytes, are known to be present in high numbers and in different developmental stages. Hence, the white body has been described as the site of hematopoiesis in cephalopods. However, to our knowledge, there are no studies showing any molecular evidence of such functions. In this study, we performed a transcriptomic analysis of white body tissue of the Southern dumpling squid, E. tasmanica. Our primary goal was to gain insights into the functions of this tissue and to test for the presence of gene transcripts associated with hematopoietic and immune processes. Several hematopoiesis genes including CPSF1, GATA 2, TFIID, and FGFR2 were found to be expressed in the white body. In addition, transcripts associated with immune-related signal transduction pathways, such as the toll-like receptor/NF-κß, and MAPK pathways were also found, as well as other immune genes previously identified in E. tasmanica's sister species, E. scolopes. This study is the first to analyze an immune organ within cephalopods, and to provide gene expression data supporting the white body as a hematopoietic tissue.


Subject(s)
Aliivibrio fischeri/immunology , Decapodiformes , Gene Expression Regulation/immunology , Hematopoiesis , Immunity/genetics , Transcriptome/immunology , Animals , Decapodiformes/genetics , Decapodiformes/immunology , Decapodiformes/metabolism , Decapodiformes/microbiology , Hematopoiesis/genetics , Hematopoiesis/immunology
5.
Elife ; 3: e01579, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24596150

ABSTRACT

Bacterial flagella mediate host-microbe interactions through tissue tropism during colonization, as well as by activating immune responses. The flagellar shaft of some bacteria, including several human pathogens, is encased in a membranous sheath of unknown function. While it has been hypothesized that the sheath may allow these bacteria to evade host responses to the immunogenic flagellin subunit, this unusual structural feature has remained an enigma. Here we demonstrate that the rotation of the sheathed flagellum in both the mutualist Vibrio fischeri and the pathogen Vibrio cholerae promotes release of a potent bacteria-derived immunogen, lipopolysaccharide, found in the flagellar sheath. We further present a new role for the flagellar sheath in triggering, rather than circumventing, host immune responses in the model squid-vibrio symbiosis. Such an observation not only has implications for the study of bacterial pathogens with sheathed flagella, but also raises important biophysical questions of sheathed-flagellum function. DOI: http://dx.doi.org/10.7554/eLife.01579.001.


Subject(s)
Aliivibrio fischeri/metabolism , Decapodiformes/microbiology , Flagella/metabolism , Lipopolysaccharides/metabolism , Vibrio cholerae/metabolism , Aliivibrio fischeri/genetics , Aliivibrio fischeri/immunology , Aliivibrio fischeri/pathogenicity , Animals , Decapodiformes/growth & development , Decapodiformes/immunology , Decapodiformes/metabolism , Flagella/immunology , Genotype , Host-Pathogen Interactions , Lipopolysaccharides/immunology , Morphogenesis , Mutation , Phenotype , Signal Transduction , Symbiosis , Vibrio cholerae/genetics , Vibrio cholerae/immunology , Vibrio cholerae/pathogenicity
6.
Semin Immunol ; 24(1): 3-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22154556

ABSTRACT

Recent research on a wide variety of systems has demonstrated that animals generally coevolve with their microbial symbionts. Although such relationships are most often established anew each generation, the partners associate with fidelity, i.e., they form exclusive alliances within the context of rich communities of non-symbiotic environmental microbes. The mechanisms by which this exclusivity is achieved and maintained remain largely unknown. Studies of the model symbiosis between the Hawaiian squid Euprymna scolopes and the marine luminous bacterium Vibrio fischeri provide evidence that the interplay between evolutionarily conserved features of the innate immune system, most notably MAMP/PRR interactions, and a specific feature of this association, i.e., luminescence, are critical for development and maintenance of this association. As such, in this partnership and perhaps others, symbiotic exclusivity is mediated by the synergism between a general animal-microbe 'language' and a 'secret language' that is decipherable only by the specific partners involved.


Subject(s)
Aliivibrio fischeri/immunology , Biological Evolution , Decapodiformes/immunology , Decapodiformes/microbiology , Symbiosis , Animals , Immunity, Innate , Receptors, Pattern Recognition/immunology
7.
Mol Microbiol ; 78(4): 903-15, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20815823

ABSTRACT

Nitric oxide (NO) is implicated in a wide range of biological processes, including innate immunity against pathogens, signal transduction and protection against oxidative stress. However, its possible roles in beneficial host-microbe associations are less well recognized. During the early stages of the squid-vibrio symbiosis, the bacterial symbiont Vibrio fischeri encounters host-derived NO, which has been hypothesized to serve as a specificity determinant. We demonstrate here that the flavohaemoglobin, Hmp, of V. fischeri protects against NO, both in culture and during colonization of the squid host. Transcriptional analyses indicate that hmp expression is highly responsive to NO, principally through the repressor, NsrR. Hmp protects V. fischeri from NO inhibition of aerobic respiration, and removes NO under both oxic and anoxic conditions. A Δhmp mutant of V. fischeri initiates squid colonization less effectively than wild type, but is rescued by the presence of an NO synthase inhibitor. The hmp promoter is activated during the initial stage of colonization, during which the Δhmp strain fails to form normal-sized aggregates of colonizing cells. Taken together, these results suggest that the sensing of host-derived NO by NsrR, and the subsequent removal of NO by Hmp, influence aggregate size and, thereby, V. fischeri colonization efficiency.


Subject(s)
Aliivibrio fischeri/immunology , Aliivibrio fischeri/physiology , Bacterial Proteins/metabolism , Decapodiformes/immunology , Decapodiformes/microbiology , Hemeproteins/metabolism , Nitric Oxide/metabolism , Symbiosis , Aerobiosis , Anaerobiosis , Animals , Gene Expression Profiling , Nitric Oxide/toxicity , Repressor Proteins/metabolism
8.
Mol Microbiol ; 77(1): 6-10, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20487269

ABSTRACT

Vibrio fischeri is a bioluminescent bacterium that enters into a symbiosis with the bobtail squid Euprymna scolopes. The bacterium colonizes a specialized light organ, in which it generates light that might help the squid to hide its silhouette from animals beneath it. Previous studies have shown that the host nitric oxide (NO) synthase is active during colonization, suggesting that V. fischeri symbionts are exposed to NO. Thus, NO might play a role in regulating the symbiosis, a role possibly analogous to that of NO in the interaction between some pathogens and their hosts. One possibility is that NO helps to exclude other species from the light organ, in which case, the response of V. fischeri to NO is of considerable interest. In this issue of Molecular Microbiology, Dunn et al. report that V. fischeri produces an NO-inducible and NO-resistant alternative oxidase (Aox) that allows respiration to continue in the presence of NO concentrations that are inhibitory to the conventional respiratory oxidases. This is an important step towards a better understanding of the role that NO plays in the Vibrio-squid symbiosis, and provides the first indication of a physiological function for a bacterial homologue of the plant Aox.


Subject(s)
Aliivibrio fischeri/physiology , Anti-Bacterial Agents/toxicity , Decapodiformes/physiology , Drug Resistance, Bacterial , Nitric Oxide/toxicity , Oxidoreductases/metabolism , Symbiosis , Aliivibrio fischeri/drug effects , Aliivibrio fischeri/immunology , Animals , Bacterial Proteins/metabolism , Decapodiformes/immunology , Decapodiformes/microbiology , Mitochondrial Proteins , Oxygen Consumption , Plant Proteins
9.
Semin Immunol ; 22(1): 48-53, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20036144

ABSTRACT

The squid-vibrio symbiosis is an experimental system being studied as a model of the chronic colonization of animal epithelia by bacterial partners. One principal question being asked with this model is: what is the role of the immune system in the dynamics of the onset and maintenance of the symbiotic state? This review focuses upon results of research to date, which have demonstrated that both cell-mediated and cell-free components of the innate immune system are involved in these processes.


Subject(s)
Aliivibrio fischeri/immunology , Decapodiformes/immunology , Immune System , Symbiosis , Aliivibrio fischeri/metabolism , Animals , Decapodiformes/metabolism , Humans , Immunity, Innate , Signal Transduction
10.
Cell Microbiol ; 11(7): 1114-27, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19416268

ABSTRACT

Peptidoglycan recognition proteins (PGRPs) are mediators of innate immunity and recently have been implicated in developmental regulation. To explore the interplay between these two roles, we characterized a PGRP in the host squid Euprymna scolopes (EsPGRP1) during colonization by the mutualistic bacterium Vibrio fischeri. Previous research on the squid-vibrio symbiosis had shown that, upon colonization of deep epithelium-lined crypts of the host light organ, symbiont-derived peptidoglycan monomers induce apoptosis-mediated regression of remote epithelial fields involved in the inoculation process. In this study, immunofluorescence microscopy revealed that EsPGRP1 localizes to the nuclei of epithelial cells, and symbiont colonization induces the loss of EsPGRP1 from apoptotic nuclei. The loss of nuclear EsPGRP1 occurred prior to DNA cleavage and breakdown of the nuclear membrane, but followed chromatin condensation, suggesting that it occurs during late-stage apoptosis. Experiments with purified peptidoglycan monomers and with V. fischeri mutants defective in peptidoglycan-monomer release provided evidence that these molecules trigger nuclear loss of EsPGRP1 and apoptosis. The demonstration of a nuclear PGRP is unprecedented, and the dynamics of EsPGRP1 during apoptosis provide a striking example of a connection between microbial recognition and developmental responses in the establishment of symbiosis.


Subject(s)
Aliivibrio fischeri/immunology , Aliivibrio fischeri/physiology , Carrier Proteins/immunology , Decapodiformes/immunology , Decapodiformes/microbiology , Peptidoglycan/immunology , Symbiosis , Aliivibrio fischeri/genetics , Amino Acid Sequence , Animals , Apoptosis , Carrier Proteins/metabolism , Cell Nucleus/chemistry , Epithelial Cells/chemistry , Epithelial Cells/microbiology , Gene Deletion , Microscopy, Fluorescence , Molecular Sequence Data , Peptidoglycan/genetics , Peptidoglycan/metabolism
11.
Environ Microbiol ; 11(2): 483-93, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19196278

ABSTRACT

The light organ crypts of the squid Euprymna scolopes permit colonization exclusively by the luminous bacterium Vibrio fischeri. Because the crypt interior remains in contact with seawater, the squid must not only foster the specific symbiosis, but also continue to exclude other bacteria. Investigation of the role of the innate immune system in these processes revealed that macrophage-like haemocytes isolated from E. scolopes recognized and phagocytosed V. fischeri less than other closely related bacterial species common to the host's environment. Interestingly, phagocytes isolated from hosts that had been cured of their symbionts bound five times more V. fischeri cells than those from uncured hosts. No such change in the ability to bind other species of bacteria was observed, suggesting that the host adapts specifically to V. fischeri. Deletion of the gene encoding OmpU, the major outer membrane protein of V. fischeri, increased binding by haemocytes from uncured animals to the level observed for haemocytes from cured animals. Co-incubation with wild-type V. fischeri reduced this binding, suggesting that they produce a factor that complements the mutant's defect. Analyses of the phagocytosis of bound cells by fluorescence-activated cell sorting indicated that once binding to haemocytes had occurred, V. fischeri cells are phagocytosed as effectively as other bacteria. Thus, discrimination by this component of the squid immune system occurs at the level of haemocyte binding, and this response: (i) is modified by previous exposure to the symbiont and (ii) relies on outer membrane and/or secreted components of the symbionts. These data suggest that regulation of host haemocyte binding by the symbiont may be one of many factors that contribute to specificity in this association.


Subject(s)
Aliivibrio fischeri/immunology , Aliivibrio fischeri/physiology , Decapodiformes/immunology , Decapodiformes/microbiology , Hemocytes/immunology , Hemocytes/microbiology , Symbiosis , Aliivibrio fischeri/genetics , Animals , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Gene Deletion , Phagocytosis
SELECTION OF CITATIONS
SEARCH DETAIL
...