Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 169
Filter
1.
Nat Commun ; 12(1): 6860, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824233

ABSTRACT

Child undernutrition is a global health issue associated with a high burden of infectious disease. Undernourished children display an overabundance of intestinal pathogens and pathobionts, and these bacteria induce enteric dysfunction in undernourished mice; however, the cause of their overgrowth remains poorly defined. Here, we show that disease-inducing human isolates of Enterobacteriaceae and Bacteroidales spp. are capable of multi-species symbiotic cross-feeding, resulting in synergistic growth of a mixed community in vitro. Growth synergy occurs uniquely under malnourished conditions limited in protein and iron: in this context, Bacteroidales spp. liberate diet- and mucin-derived sugars and Enterobacteriaceae spp. enhance the bioavailability of iron. Analysis of human microbiota datasets reveals that Bacteroidaceae and Enterobacteriaceae are strongly correlated in undernourished children, but not in adequately nourished children, consistent with a diet-dependent growth synergy in the human gut. Together these data suggest that dietary cross-feeding fuels the overgrowth of pathobionts in undernutrition.


Subject(s)
Gastrointestinal Microbiome/physiology , Malnutrition/microbiology , Animals , Bacteroidetes/growth & development , Bacteroidetes/isolation & purification , Bacteroidetes/metabolism , Child , Coculture Techniques , Diet/adverse effects , Enterobacteriaceae/growth & development , Enterobacteriaceae/isolation & purification , Enterobacteriaceae/metabolism , Escherichia coli/growth & development , Escherichia coli/isolation & purification , Escherichia coli/metabolism , Feces/microbiology , Humans , Intestines/chemistry , Intestines/microbiology , Mice , Nutrients/analysis , Nutrients/metabolism , Symbiosis
2.
Mucosal Immunol ; 4(1): 8-14, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21150896

ABSTRACT

The human interface with the microbial world has so far largely been considered through the somewhat restrictive angle of host-pathogen interactions resulting in disease. It has consequently largely ignored the daily symbiosis with the microbiota, an ensemble of symbiotic microorganisms engaged in a commensal, and for some of them mutualistic, interaction. This microbiota heavily populates essential surfaces such as the oral and intestinal cavity, the upper respiratory tract, the vagina, and the skin. Host response to the pathogens is characterized by quick recognition combined with strong innate (i.e., inflammatory) and adaptive immune responses, causing microbial eradication often at the cost of significant tissue damage. Response to the symbiotic microbiota is characterized by a process called tolerance that encompasses a complex integration of microbial recognition and tightly controlled innate (i.e., physiological inflammation) and adaptive immune responses. This dichotomy in host response is critical at the gut mucosal surface that is massively colonized by a diverse population of bacteria. The host is therefore permanently facing the challenge of discriminating among symbiotic and pathogenic bacteria in order to offer an adapted response. This asks the fundamental existential question: "to be or not to be… a pathogen." This review has attempted to consider this question from the host angle. What do host mucosal sensing systems see in the bacteria to which they become exposed to establish proper discrimination? A new facet of medicine resides in the dysfunction of this complex balance that has likely forged the complexity of the immune system.


Subject(s)
Host-Pathogen Interactions , Immunity, Mucosal , Intestinal Mucosa/microbiology , Mucous Membrane/microbiology , Receptors, Pattern Recognition/immunology , Symbiosis , Adaptive Immunity , Biomass , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Humans , Immune Tolerance , Immunity, Innate , Inflammation/immunology , Intestinal Mucosa/immunology , Mucous Membrane/immunology , Receptors, Pattern Recognition/genetics , Respiratory System/immunology , Respiratory System/microbiology , Skin/immunology , Skin/microbiology
3.
Mol Immunol ; 46(13): 2647-54, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19535145

ABSTRACT

The WD40 repeat containing angio-associated migratory cell protein (AAMP) was identified as a new binding partner of the human nucleotide-binding domain, leucine rich repeat containing (NLR) family member Nod2 in a yeast two-hybrid screen. Co-immunoprecipitations from human cells verified this interaction and revealed that an internal peptide of AAMP spanning three WD40 domains was sufficient for this interaction. AAMP was found to be ubiquitously expressed in different human cell-lines and exhibited a predominant cytosolic localization in epithelial cells. Functionally, using overexpression and siRNA knock-down, we showed that AAMP modulates Nod2- and Nod1-mediated NF-kappaB activation in HEK293T cells. Taken together, our data support a new function of AAMP in regulating innate immune responses initiated by the NLR protein Nod2.


Subject(s)
Carrier Proteins/metabolism , NF-kappa B/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Receptors, Antigen, B-Cell/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Carrier Proteins/genetics , Cell Line , HeLa Cells , Humans , Immunoblotting , Luciferases/genetics , Luciferases/metabolism , Microscopy, Fluorescence , Molecular Sequence Data , Nod1 Signaling Adaptor Protein/genetics , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Plasmids/genetics , Protein Binding , Protein Interaction Domains and Motifs/genetics , Protein Interaction Mapping , Receptors, Antigen, B-Cell/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Signal Transduction
4.
Cell Microbiol ; 5(7): 481-91, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12814438

ABSTRACT

Shigella infection is characterized by the induction of acute inflammation, which is responsible for the massive tissue destruction of the intestinal mucosa. A murine model would be a valuable tool for gaining a better understanding of the physiopathology of shigellosis and the host immune response to Shigella infection, but adult mice do not develop disease upon oral inoculation. We therefore attempted to develop a model of infection in newborn mice. Four-day-old mice inoculated with 50 microl of 5 x 10(9) invasive wild-type Shigella flexneri 5a were susceptible to bacterial infection, but mice inoculated with the non-invasive strain BS176 were not. Histologically, 4-day-old mice infected with the invasive strain presented intestinal lesions and inflammation similar to those described in patients with shigellosis. Moreover, cytokine and chemokine responses consistent with inflammation were observed. Lower bacterial inocula induced less severe intestinal damage. In contrast, 5-day-old mice inoculated with either the invasive or the non-invasive strain were not infected. We have thus established a mouse model that is suitable for the study of the pathogenesis of intestinal Shigella infection.


Subject(s)
Disease Models, Animal , Dysentery, Bacillary/physiopathology , Shigella flexneri/pathogenicity , Animals , Animals, Newborn , Cytokines/biosynthesis , Cytokines/genetics , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/pathology , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred BALB C , Microscopy, Confocal
5.
Crit Rev Immunol ; 23(5-6): 371-401, 2003.
Article in English | MEDLINE | ID: mdl-15030306

ABSTRACT

Acute infectious colitis remains a major pediatric issue of worldwide impact because it still represents a significant public health burden among the larger group of diarrheal diseases with the highest mortality rate. It is also a relevant model of inflammatory bowel diseases (IBD), such as Crohn's disease and ulcerative colitis. Among cases of acute colitis of infectious origin, shigellosis is certainly the one that has benefited the most from a significant research effort. Shigella, the causative agent, is a Gram-negative bacterium that has the capacity to invade, disrupt, and cause inflammatory destruction of the intestinal epithelial barrier. The molecular and cellular bases of this invasive phenotype essentially encompass crossing of the epithelial lining, apoptotic killing of macrophages, entry into epithelial cells, and escape into the cytoplasm, followed by cell-to-cell spread. Intracellular colonization is likely to protect the micro-organisms from killing by humoral and cellular effectors of the innate immune response. Concurrently, the capacity of Shigella to reprogram invaded epithelial cells to produce proinflammatory mediators plays a major role in the strong inflammatory profile of the disease. This profile is likely to impact on the nature and quality of the adaptive response, which is dominated by humoral protection at the mucosal level.


Subject(s)
Dysentery, Bacillary/immunology , Shigella Vaccines/isolation & purification , Shigella/immunology , Shigella/pathogenicity , Adaptation, Physiological , Animals , Bacterial Proteins/immunology , Dendritic Cells/microbiology , Dysentery, Bacillary/etiology , Dysentery, Bacillary/pathology , Dysentery, Bacillary/prevention & control , Epithelial Cells/immunology , Epithelial Cells/pathology , Humans , Immunity, Innate , Immunity, Mucosal , In Vitro Techniques , Intestines/immunology , Intestines/pathology , Macrophages/microbiology
6.
Curr Opin Immunol ; 13(4): 410-6, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11498296

ABSTRACT

The ability to discriminate between pathogenic and non-pathogenic bacteria is extremely important for epithelial cells lining mucosal surfaces and is particularly so in colonic epithelial cells. Accumulating evidence suggests that bacterial recognition systems used by epithelial cells are very different from those in cells of the myeloid lineage and are likely to have developed to maintain mucosal surfaces in a state of homeostasis with the normal microbial flora. Bacterial invasion of epithelial cells or breach of the epithelial barrier provides a signal to epithelial cells to initiate inflammatory responses, which are key events for the clearance of the infecting microbe. Therefore, elucidation of the mechanisms by which epithelial cells recognize bacteria and bacterial products, and of the nature of the innate immune responses that are triggered by these factors are important for our understanding of both the immunology of mucosal surfaces and bacterial pathogenesis.


Subject(s)
Bacterial Infections/immunology , Animals , Apoptosis , Bacteria/immunology , Bacteria/pathogenicity , Bacterial Infections/genetics , Bacterial Infections/microbiology , Bacterial Infections/pathology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Gene Expression , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Models, Biological , Signal Transduction
7.
Infect Immun ; 69(9): 5230-4, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11500390

ABSTRACT

The mucosal humoral immune response elicited following Shigella flexneri infection in patients living in Antananarivo districts (Madagascar Island) was evaluated by measuring the gut-derived, circulating immunoglobulin A (IgA) antibody-secreting cells (ASC) specific for the major bacterial antigen lipopolysaccharide (LPS). Fifty, 34, 11, and 5% of the S. flexneri-positive patients were infected with serotypes 2a, 1a, 4a, and 3a, respectively. The total number of IgA ASC in infected patients increased significantly, compared to the number in healthy controls, early after the onset of disease. The number of anti-homologous LPS IgA ASC varied among individuals and peaked between days 5 and 10 after the onset of the disease. In the S. flexneri 1a- and 2a-infected patients, the level of IgA ASC cross-reactivity to heterologous S. flexneri serotypes was weak. These data indicate that S. flexneri 2a and 1a are the predominant strains responsible for shigellosis in this area of endemicity and that the anti-LPS antibody response following natural infection is mainly directed against serotype-specific determinants.


Subject(s)
Antibody-Producing Cells/immunology , Dysentery, Bacillary/immunology , Immunity, Mucosal , Immunoglobulin A, Secretory/blood , Lipopolysaccharides/immunology , Shigella flexneri/immunology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Child , Child, Preschool , Cross Reactions , Dysentery, Bacillary/epidemiology , Dysentery, Bacillary/microbiology , Endemic Diseases , Humans , Immunoglobulin A, Secretory/immunology , Intestines/immunology , Prevalence , Serotyping , Shigella flexneri/classification
8.
EMBO Rep ; 2(8): 736-42, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11463746

ABSTRACT

Epithelial cells are refractory to extracellular lipopolysaccharide (LPS), yet when presented inside the cell, it is capable of initiating an inflammatory response. Using invasive Shigella flexneri to deliver LPS into the cytosol, we examined how this factor, once intracellular, activates both NF-kappaB and c-Jun N-terminal kinase (JNK). Surprisingly, the mode of activation is distinct from that induced by toll-like receptors (TLRs), which mediate LPS responsiveness from the outside-in. Instead, our findings demonstrate that this response is mediated by a cytosolic, plant disease resistance-like protein called CARD4/Nod1. Biochemical studies reveal enhanced oligomerization of CARD4 upon S. flexneri infection, an event necessary for NF-kappaB induction. Dominant-negative versions of CARD4 block activation of NF-kappaB and JNK by S. flexneri as well as microinjected LPS. Finally, we showed that invasive S. flexneri triggers the formation of a transient complex involving CARD4, RICK and the IKK complex. This study demonstrates that in addition to the extracellular LPS sensing system mediated by TLRs, mammalian cells also possess a cytoplasmic means of LPS detection via a molecule that is related to plant disease-resistance proteins.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Drosophila Proteins , Gene Expression Regulation/physiology , Lipopolysaccharides/pharmacology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Shigella flexneri/physiology , Signal Transduction/physiology , Carrier Proteins/genetics , Cell Line , Genes, Reporter , HeLa Cells , Humans , I-kappa B Kinase , Interleukin-1/pharmacology , JNK Mitogen-Activated Protein Kinases , Lipopolysaccharides/administration & dosage , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microinjections , Nod1 Signaling Adaptor Protein , Precipitin Tests , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteins/genetics , Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Shigella flexneri/pathogenicity , TNF Receptor-Associated Factor 2 , Toll-Like Receptors , Tumor Necrosis Factor-alpha/pharmacology
9.
Infect Immun ; 69(2): 1072-83, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160004

ABSTRACT

Because the use of live attenuated mutants of Shigella spp. represents a promising approach to protection against bacillary dysentery (M. E. Etherridge, A. T. M. Shamsul Hoque, and D. A. Sack, Lab. Anim. Sci. 46:61-66, 1996), it becomes essential to rationalize this approach in animal models in order to optimize attenuation of virulence in the vaccine candidates, as well as their route and mode of administration, and to define the correlates of protection. In this study, we have compared three strains of Shigella flexneri 5--the wild-type M90T, an aroC mutant, and a double purE aroC mutant--for their pathogenicity, immunogenicity, and protective capacity. Protection against keratoconjunctivitis, induced by wild-type M90T, was used as the protection read out in guinea pigs that were inoculated either intranasally or intragastrically. Following intranasal immunization, the aroC mutant elicited weak nasal tissue destruction compared to M90T and achieved protection correlated with high levels of local anti-lipopolysaccharide immunoglobulin A (IgA), whereas the purE aroC double mutant, which also elicited weak tissue destruction, was not protective and elicited a low IgA response. Conversely, following intragastric immunization, only the M90T purE aroC double mutant elicited protection compared to both the aroC mutant and the wild-type strain. This mutant caused mild inflammatory destruction, particularly at the level of Peyer's patches, but it persisted much longer within the tissues. This could represent an essential parameter of the protective response that, in this case, did not clearly correlate with high anti-lipopolysaccharide IgA titers.


Subject(s)
Bacterial Vaccines/immunology , Dysentery, Bacillary/prevention & control , Shigella flexneri/immunology , Animals , Antibodies, Bacterial/blood , Dysentery, Bacillary/metabolism , Dysentery, Bacillary/pathology , Female , Guinea Pigs , Immunization , Immunohistochemistry , Interferon-gamma/biosynthesis , Lipopolysaccharides/analysis , Lipopolysaccharides/immunology , Mutation , Vaccines, Attenuated/immunology
10.
Am J Physiol Gastrointest Liver Physiol ; 280(3): G319-23, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11171613

ABSTRACT

Interaction of Shigella flexneri with epithelial cells includes contact of bacteria with the cell surface and release of Ipa proteins through a specialized type III secreton. A complex signaling process involving activation of small GTPases of the Rho family and c-src causes major rearrangements of the subcortical cytoskeleton, thereby allowing bacterial entry by macropinocytosis. After entry, shigellae escape to the cell cytoplasm and initiate intracytoplasmic movement through polar nucleation and assembly of actin filaments caused by bacterial surface protein IcsA, which binds and activates neuronal Wiskoff-Aldrich syndrome protein (N-WASP), thus inducing actin nucleation in an Arp 2/3-dependent mechanism. Actin-driven motility promotes efficient colonization of the host cell cytoplasm and rapid cell-to-cell spread via protrusions that are engulfed by adjacent cells in a cadherin-dependent process. Bacterial invasion turns infected cells to strongly proinflammatory cells through sustained activation of nuclear factor-kappaB. A major consequence is interleukin (IL)-8 production, which attracts polymorphonuclear leukocytes (PMNs). On transmigration, PMNs disrupt the permeability of this epithelium and promote its invasion by shigellae. At the early stage of infection, M cells of the follicle-associated epithelium allow bacterial translocation. Subsequent apoptotic killing of macrophages in a caspase 1-dependent process causes the release of IL-1beta and IL-18, which accounts for the initial steps of inflammation.


Subject(s)
Dysentery, Bacillary/immunology , Dysentery, Bacillary/microbiology , Host-Parasite Interactions/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Dysentery, Bacillary/pathology , Humans , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Intestinal Mucosa/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/microbiology , Shigella flexneri/immunology , Shigella flexneri/pathogenicity , Toxins, Biological/toxicity
12.
Mol Microbiol ; 38(1): 8-19, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11029686

ABSTRACT

Invasion of epithelial cells by Shigella flexneri involves entry and intercellular dissemination. Entry of bacteria into non-phagocytic cells requires the IpaA-D proteins that are secreted by the Mxi-Spa type III secretion machinery. Type III secretion systems are found in several Gram-negative pathogens and serve to inject bacterial effector proteins directly into the cytoplasm of host cells. In this study, we have analysed the IpgD protein of S. flexneri, the gene of which is located on the virulence plasmid at the 5' end of the mxi-spa locus. We have shown that IpgD (i) is stored in the bacterial cytoplasm in association with a specific chaperone, IpgE; (ii) is secreted by the Mxi-Spa type III secretion system in amounts similar to those of the IpaA-D proteins; (iii) is associated with IpaA in the extracellular medium; and (iv) is involved in the modulation of the host cell response after contact of the bacterium with epithelial cells. This suggests that IpgD is an effector that might be injected into host cells to manipulate cellular processes during infection.


Subject(s)
Bacterial Proteins/metabolism , Membrane Fusion , Molecular Chaperones/metabolism , Shigella flexneri/physiology , Base Sequence , Culture Media , DNA Primers , HeLa Cells , Humans
13.
Microbes Infect ; 2(7): 813-9, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10955962

ABSTRACT

Shigella, the causative agent of bacillar dysentery, invades colonic epithelial cells and moves intracellularly to spread from cell to cell. The processes of Shigella entry, determined by the Ipa proteins, and of actin-based motility, dependent on the IcsA/VirG protein, represent different levels of bacterial manipulation of the cell cytoskeleton.


Subject(s)
Cytoskeleton/metabolism , Epithelial Cells/microbiology , Intestinal Mucosa/microbiology , Shigella/pathogenicity , Actins/metabolism , Animals , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , DNA-Binding Proteins/metabolism , Humans , Intestinal Mucosa/cytology , Nerve Tissue Proteins/metabolism , Shigella/metabolism , Shigella/physiology , Transcription Factors/metabolism , Vinculin/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal , rho GTP-Binding Proteins/metabolism , src-Family Kinases/metabolism
14.
J Immunol ; 165(2): 903-14, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10878365

ABSTRACT

The pathogenesis of Shigella flexneri infection centers on the ability of this organism to invade epithelial cells and initiate an intense inflammatory reaction. Because NF-kappa B is an important transcriptional regulator of genes involved in inflammation, we investigated the role of this transcription factor during S. flexneri infection of epithelial cells. Infection of HeLa cells with invasive S. flexneri induced NF-kappa B DNA-binding activity; noninvasive S. flexneri strains did not lead to this activation. The pathway leading to NF-kappa B activation by invasive S. flexneri involved the kinases, NF-kappa B-inducing kinase, I kappa B kinase-1, and I kappa B kinase-2. NF-kappa B activation was linked to inflammation, because invasive S. flexneri activated an IL-8 promoter-driven reporter gene, and the kappa B site within this promoter was indispensable for its induction. Microinjection of bacterial culture supernatants into HeLa cells suggested that LPS is responsible for NF-kappa B activation by S. flexneri infection. In conclusion, the eukaryotic transcription factor NF-kappa B was activated during S. flexneri infection of epithelial cells, which suggests a role for this transcriptional regulator in modulating the immune response during infection in vivo.


Subject(s)
Epithelial Cells/immunology , Epithelial Cells/metabolism , Interleukin-8/biosynthesis , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Shigella flexneri/immunology , Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Binding Sites/genetics , Binding Sites/immunology , Caco-2 Cells , DNA-Binding Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/immunology , Epithelial Cells/enzymology , Epithelial Cells/microbiology , HeLa Cells , Humans , I-kappa B Kinase , Immunity, Innate , Interleukin-8/genetics , Interleukin-8/metabolism , Intracellular Fluid/microbiology , Luciferases/biosynthesis , Luciferases/genetics , Luciferases/metabolism , NF-kappa B/biosynthesis , Promoter Regions, Genetic/immunology , Protein Serine-Threonine Kinases/metabolism , Shigella flexneri/pathogenicity , Signal Transduction/immunology , Virulence/immunology
15.
Immunity ; 12(5): 581-90, 2000 May.
Article in English | MEDLINE | ID: mdl-10843390

ABSTRACT

Caspases are intracellular proteases that mediate mammalian cell apoptosis. Caspase-1 (Casp-1) is a unique caspase because it activates the proinflammatory cytokines interleukin (IL)-1beta and IL-18. Shigella flexneri, the etiological agent of bacillary dysentery, induces macrophage apoptosis, which requires Casp-1 and results in the release of mature IL-1beta and IL-18. Here we show that casp-1(-/-) mice infected with S. flexneri do not develop the acute inflammation characteristic of shigellosis and are unable to resolve the bacterial infection. Using casp-1(-/-) mice supplemented with recombinant cytokines and experiments with IL-1beta(-/-) and IL-18(-/-) mice, we show that IL-1beta and IL-18 are both required to mediate inflammation in S. flexneri infections. Together, these data demonstrate the importance of Casp-1 in acute inflammation and show the different roles of its substrates, IL-1beta and IL-18, in this response.


Subject(s)
Caspase 1/immunology , Dysentery, Bacillary/immunology , Interleukin-18/immunology , Interleukin-1/immunology , Shigella flexneri , Animals , Dysentery, Bacillary/metabolism , Immunohistochemistry , Inflammation/immunology , Mice , Substrate Specificity/immunology
16.
Philos Trans R Soc Lond B Biol Sci ; 355(1397): 575-86, 2000 May 29.
Article in English | MEDLINE | ID: mdl-10874731

ABSTRACT

Shigella flexneri is a Gram-negative facultatively intracellular pathogen responsible for bacillary dysentery in humans. More than one million deaths occur yearly due to infections with Shigella spp. and the victims are mostly children of the developing world. The pathogenesis of Shigella centres on the ability of this organism to invade the colonic epithelium where it induces severe mucosal inflammation. Much information that we have gained concerning the pathogenesis of Shigella has been derived from the study of in vitro models of infection. Using these techniques, a number of the molecular mechanisms by which Shigella invades epithelial cells and macrophages have been identified. In vivo models of shigellosis have been hampered since humans are the only natural hosts of Shigella. However, experimental infection of macaques as well as the murine lung and rabbit ligated ileal loop models have been important in defining some of the immune and inflammatory components of the disease. In particular, the murine lung model has shed light on the development of systemic and local immune protection against Shigella infection. It would be naive to believe that any one model of Shigella infection could adequately represent the complexity of the disease in humans, and more sophisticated in vivo models are now necessary. These models require the use of human cells and tissue, but at present such models remain in the developmental stage. Ultimately, however, it is with such studies that novel treatments and vaccine candidates for the treatment and prevention of shigellosis will be designed.


Subject(s)
Dysentery, Bacillary/microbiology , Shigella flexneri/pathogenicity , Adaptation, Physiological/immunology , Animals , Apoptosis , Cell Adhesion , Dysentery, Bacillary/immunology , Epithelial Cells/microbiology , Humans , Intracellular Fluid/microbiology , Macrophages/microbiology , Rabbits , Receptors, Cell Surface/metabolism , Shigella flexneri/immunology
19.
Infect Immun ; 68(3): 1289-96, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10678940

ABSTRACT

Infection by Shigella flexneri is characterized by infiltration of neutrophils in the intestinal mucosa and by a strong inflammatory reaction. Although neutrophils are constitutively programmed to die by apoptosis, we show that isolated human neutrophils undergo necrosis 2 h after infection with virulent S. flexneri strain M90T but not with the virulence plasmid-cured strain BS176. This was demonstrated by the release of azurophil granule proteins concomitant with the release of lactate dehydrogenase (LDH), disruption of the plasma membrane, and absence of DNA fragmentation. Mutants with the mxiD1 gene, coding for an essential component of the secretion type III machinery, or the genes coding for IpaB or IpaC invasins deleted were not cytotoxic. Neutrophil necrosis occurred independently of the bacterial ability to leave phagosomes, and it involved actin polymerization, as the addition of cytochalasin D after phagocytosis of Shigella inhibited the release of LDH. In conclusion, Shigella kills neutrophils by necrosis, a process characterized by the release of tissue-injurious granular proteins. This probably contributes to disruption of the epithelial barrier, leading to the dysentery observed in shigellosis and allowing Shigella to enter its host cells.


Subject(s)
Actins/metabolism , Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Neutrophils/pathology , Shigella flexneri/immunology , Apoptosis , Exocytosis , Humans , Lysosomes/physiology , Membrane Fusion , Necrosis , Phagocytosis , Polymers/metabolism , Vacuoles/microbiology
20.
J Immunol ; 164(4): 2221-32, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657678

ABSTRACT

Volunteers were orally administered invasive, non-Shiga toxin-producing Shigella dysenteriae 1 to establish a challenge model to assess vaccine efficacy. In stepwise fashion, four separate groups were given 3 x 10(2), 7 x 10(3), 5 x 10(4), or 7 x 10(5) CFU. Using PBMC, proliferative responses and cytokine production were measured to S. dysenteriae whole-cell preparations and to purified recombinant invasion plasmid Ags (Ipa) C and IpaD. Anti-LPS and anti-Ipa Abs and Ab-secreting cells were also evaluated. Preinoculation PBMC produced considerable quantities of IL-10 and IFN-gamma, probably secreted by monocytes and NK cells, respectively, of the innate immune system. Following inoculation, PBMC from 95 and 87% of volunteers exhibited an increased production of IFN-gamma and IL-10, respectively, in response to Shigella Ags. These increases included responses to IpaC and IpaD among those volunteers receiving the lowest inoculum. No IL-4 or IL-5 responses were detected. Whereas there were no Ab or Ab-secreting cell responses in volunteers receiving the lowest inoculum, other dose groups had moderate to strong anti-LPS and anti-Ipa responses. These results suggest that in humans, type 1 responses play an important role in mucosal and systemic immunity to S. dysentariae 1.


Subject(s)
Adhesins, Bacterial , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Bacterial Vaccines/immunology , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Leukocytes, Mononuclear/immunology , Shigella dysenteriae/immunology , Vaccines, Synthetic/immunology , Administration, Oral , Adolescent , Adult , Antibodies, Bacterial/biosynthesis , Antibody-Producing Cells/immunology , Antibody-Producing Cells/metabolism , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Toxins/administration & dosage , Bacterial Toxins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Colony Count, Microbial , Dose-Response Relationship, Immunologic , Dysentery, Bacillary/immunology , Dysentery, Bacillary/metabolism , Dysentery, Bacillary/prevention & control , Gene Deletion , Humans , Interleukin-12/biosynthesis , Interleukin-15/biosynthesis , Interleukin-2/biosynthesis , Interleukin-4/biosynthesis , Interleukin-5/biosynthesis , Kinetics , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/microbiology , Lymphocyte Activation , Shiga Toxins , Shigella dysenteriae/genetics , Transforming Growth Factor beta/biosynthesis , Vaccines, Synthetic/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...