Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Crohns Colitis ; 15(9): 1596-1601, 2021 Sep 25.
Article in English | MEDLINE | ID: mdl-33631789

ABSTRACT

BACKGROUND AND AIMS: Antibodies targeting tumor necrosis factor-alpha [TNF-alpha] are a mainstay in the treatment of inflammatory bowel disease. However, they fail to demonstrate efficacy in a considerable proportion of patients. On the other hand, glycosylation of antibodies might influence not only their immunogenicity but also their structure and function. We investigated whether specific glycosylation patterns of the Fc-fragment would affect the immunogenicity of anti-TNF-alpha antibody in monocyte-derived dendritic cells. METHODS: The effect of a specific Fc-glycosylation pattern on antibody uptake by monocyte-derived dendritic cells [mo-DCs] and how this process shapes the immunologic profile of mo-DCs was investigated. Three N-glycoforms of the anti-TNF-alpha antibody adalimumab, that differed in the content of fucose or sialic acid, were tested: [1] mock treated Humira, abbreviated 'Fuc-G0', where the N-glycan mainly consist of fucose and N-acetylglucosamine [GlcNAc], without sialic acid; [2] 'Fuc-G2S1/G2S2' with fucose and alpha 2,6 linked sialic acid; and [3] 'G2S1/G2S2' with alpha 2,6 linked sialic acid, without fucose. RESULTS: Our data demonstrated that neither fucosylation nor sialylation of anti-TNF-Abs [Fuc-G0, FucG2S1/G2S2, G2S1/G2S2] influence their uptake by mo-DCs. Additionally, none of the differentially glycosylated antibodies altered CD80, CD86, CD273, CD274 levels on mo-DCs stimulated in with lipopolysaccharide in the presence of antibodies. Next, we evaluated the levels of cytokines in the supernatant of mo-DCs stimulated with lipopolysaccharide in the presence of Fuc-G0, Fuc-G2S1/G2S2 or G2S1/G2S2-glycosylated anti-TNF antibodies. Only IL-2 and IL-17 levels were downregulated, and IL-5 production was upregulated by uptake of Fuc-G0 antibodies, as compared to control without antibodies. CONCLUSIONS: The specific modification in the Fc-glycosylation pattern of anti-TNF-alpha Abs does not affect their immunogenicity under the tested conditions. As this study was limited to mo-DCs, further investigation is required to clarify whether Ab uptake into mo-DCs might change the immunological profile of T- and B-cells, in order to ultimately reduce the formation of anti-drug antibodies and to improve the patient care.


Subject(s)
Adalimumab/pharmacology , Dendritic Cells/drug effects , Fucose/metabolism , Immunoglobulin Fc Fragments/metabolism , N-Acetylneuraminic Acid/metabolism , Tumor Necrosis Factor Inhibitors/pharmacology , Adalimumab/chemistry , Cell Culture Techniques , Glycosylation , Humans , Monocytes , Tumor Necrosis Factor Inhibitors/chemistry
2.
Metab Eng ; 44: 293-301, 2017 11.
Article in English | MEDLINE | ID: mdl-29101090

ABSTRACT

Polysialic acid (polySia) is a posttranslational modification found on only a handful of proteins in the central nervous and immune systems. The addition of polySia to therapeutic proteins improves pharmacokinetics and reduces immunogenicity. To date, polysialylation of therapeutic proteins has only been achieved in vitro by chemical or chemoenzymatic strategies. In this work, we develop a biosynthetic pathway for site-specific polysialylation of recombinant proteins in the cytoplasm of Escherichia coli. The pathway takes advantage of a bacterial cytoplasmic polypeptide-glycosyltransferase to establish a site-specific primer on the target protein. The glucose primer is extended by glycosyltransferases derived from lipooligosaccharide, lipopolysaccharide and capsular polysaccharide biosynthesis from different bacterial species to synthesize long chain polySia. We demonstrate the new biosynthetic route by modifying green fluorescent proteins and a therapeutic DARPin (designed ankyrin repeat protein).


Subject(s)
Escherichia coli , Protein Modification, Translational/genetics , Sialic Acids , Escherichia coli/genetics , Escherichia coli/metabolism , Glycosylation , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Sialic Acids/genetics , Sialic Acids/metabolism
3.
Sci Rep ; 6: 38914, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27974829

ABSTRACT

Capnocytophaga canimorsus is a dog's and cat's oral commensal which can cause fatal human infections upon bites or scratches. Infections mainly start with flu-like symptoms but can rapidly evolve in fatal septicaemia with a mortality as high as 40%. Here we present the discovery of a polysaccharide capsule (CPS) at the surface of C. canimorsus 5 (Cc5), a strain isolated from a fulminant septicaemia. We provide genetic and chemical data showing that this capsule is related to the lipooligosaccharide (LOS) and probably composed of the same polysaccharide units. A CPS was also found in nine out of nine other strains of C. canimorsus. In addition, the genomes of three of these strains, sequenced previously, contain genes similar to those encoding CPS biosynthesis in Cc5. Thus, the presence of a CPS is likely to be a common property of C. canimorsus. The CPS and not the LOS confers protection against the bactericidal effect of human serum and phagocytosis by macrophages. An antiserum raised against the capsule increased the killing of C. canimorsus by human serum thus showing that anti-capsule antibodies have a protective role. These findings provide a new major element in the understanding of the pathogenesis of C. canimorsus.


Subject(s)
Bacterial Capsules/chemistry , Capnocytophaga/chemistry , Lipopolysaccharides/chemistry , Polysaccharides, Bacterial/chemistry , Animals , Antibodies, Bacterial/immunology , Bacterial Capsules/immunology , Capnocytophaga/immunology , Capnocytophaga/pathogenicity , Cats , Dogs , Gram-Negative Bacterial Infections/immunology , Humans , Lipopolysaccharides/immunology , Polysaccharides, Bacterial/immunology
5.
Mol Microbiol ; 87(1): 112-31, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23163552

ABSTRACT

Bacterial carbohydrate structures play a central role in mediating a variety of host-pathogen interactions. Glycans can either elicit protective immune response or lead to escape of immune surveillance by mimicking host structures. Lipopolysaccharide (LPS), a major component on the surface of Gram-negative bacteria, is composed of a lipid A-core and the O-antigen polysaccharide. Pathogens like Neisseria meningitidis expose a lipooligosaccharide (LOS), which outermost glycans mimick mammalian epitopes to avoid immune recognition. Lewis X (Galß1-4(Fucα1-3)GlcNAc) antigens of Helicobacter pylori or of the helminth Schistosoma mansoni modulate the immune response by interacting with receptors on human dendritic cells. In a glycoengineering approach we generate human carbohydrate structures on the surface of recombinant Gram-negative bacteria, such as Escherichia coli and Salmonella enterica sv. Typhimurium that lack O-antigen. A ubiquitous building block in mammalian N-linked protein glycans is Galß1-4GlcNAc, referred to as a type-2 N-acetyllactosamine, LacNAc, sequence. Strains displaying polymeric LacNAc were generated by introducing a combination of glycosyltransferases that act on modified lipid A-cores, resulting in efficient expression of the carbohydrate epitope on bacterial cell surfaces. The poly-LacNAc scaffold was used as an acceptor for fucosylation leading to polymers of Lewis X antigens. We analysed the distribution of the carbohydrate epitopes by FACS, microscopy and ELISA and confirmed engineered LOS containing LacNAc and Lewis X repeats by MALDI-TOF and NMR analysis. Glycoengineered LOS induced pro-inflammatory response in murine dendritic cells. These bacterial strains can thus serve as tools to analyse the role of defined carbohydrate structures in different biological processes.


Subject(s)
Antigens, Surface/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Lewis X Antigen/genetics , Lewis X Antigen/metabolism , Animals , Dendritic Cells/immunology , Dendritic Cells/microbiology , Disaccharides/immunology , Escherichia coli/metabolism , Helicobacter pylori/metabolism , Host-Pathogen Interactions , Lewis X Antigen/immunology , Lipopolysaccharides/immunology , Mice , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Salmonella typhimurium/metabolism , Schistosoma mansoni/metabolism
6.
PLoS Pathog ; 7(6): e1002118, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21738475

ABSTRACT

C. canimorsus 5 has the capacity to grow at the expenses of glycan moieties from host cells N-glycoproteins. Here, we show that C. canimorsus 5 also has the capacity to deglycosylate human IgG and we analyze the deglycosylation mechanism. We show that deglycosylation is achieved by a large complex spanning the outer membrane and consisting of the Gpd proteins and sialidase SiaC. GpdD, -G, -E and -F are surface-exposed outer membrane lipoproteins. GpdDEF could contribute to the binding of glycoproteins at the bacterial surface while GpdG is a endo-ß-N-acetylglucosaminidase cleaving the N-linked oligosaccharide after the first N-linked GlcNAc residue. GpdC, resembling a TonB-dependent OM transporter is presumed to import the oligosaccharide into the periplasm after its cleavage from the glycoprotein. The terminal sialic acid residue of the oligosaccharide is then removed by SiaC, a periplasm-exposed lipoprotein in direct contact with GpdC. Finally, most likely degradation of the oligosaccharide proceeds sequentially from the desialylated non reducing end by the action of periplasmic exoglycosidases, including ß-galactosidases, ß-N-Acetylhexosaminidases and α-mannosidases.


Subject(s)
Capnocytophaga/metabolism , Glycoproteins/metabolism , Immunoglobulin G/metabolism , Bacterial Outer Membrane Proteins/metabolism , Cell Line , Glycosylation , Gram-Negative Bacterial Infections , Humans , Lipoproteins/metabolism , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/metabolism , N-Acetylneuraminic Acid/metabolism , Neuraminidase/metabolism , Polysaccharides/chemistry , Polysaccharides/metabolism , alpha-Mannosidosis/metabolism , beta-Galactosidase/metabolism , beta-N-Acetylhexosaminidases/metabolism
7.
Mol Microbiol ; 81(4): 1050-60, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21762219

ABSTRACT

Capnocytophaga canimorsus are commensal Gram-negative bacteria from dog's mouth that cause rare but dramatic septicaemia in humans. C. canimorsus have the unusual property to feed on cultured mammalian cells, including phagocytes, by harvesting the glycan moiety of cellular glycoproteins. To understand the mechanism behind this unusual property, the genome of strain Cc5 was sequenced and analysed. In addition, Cc5 bacteria were cultivated onto HEK 293 cells and the surface proteome was determined. The genome was found to encode many lipoproteins encoded within 13 polysaccharide utilization loci (PULs) typical of the Flavobacteria-Bacteroides group. PULs encode surface exposed feeding complexes resembling the archetypal starch utilization system (Sus). The products of at least nine PULs were detected among the surface proteome and eight of them represented more than half of the total peptides detected from the surface proteome. Systematic deletions of the 13 PULs revealed that half of these Sus-like complexes contributed to growth on animal cells. The complex encoded by PUL5, one of the most abundant ones, was involved in foraging glycans from glycoproteins. It was essential for growth on cells and contributed to survival in mice. It thus represents a fitness factor during infection.


Subject(s)
Bacterial Proteins/metabolism , Capnocytophaga/metabolism , Glycoproteins/metabolism , Polysaccharides/metabolism , Proteome/metabolism , Animals , Cell Line , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Epithelial Cells/microbiology , Genes, Bacterial , Genome, Bacterial , Humans , Metabolic Networks and Pathways/genetics , Mice , Molecular Sequence Data , Multigene Family , Sequence Analysis, DNA
8.
Infect Immun ; 77(6): 2262-71, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19307219

ABSTRACT

Capnocytophaga canimorsus is a bacterium of the canine oral flora known since 1976 to cause rare but severe septicemia and peripheral gangrene in patients that have been in contact with a dog. It was recently shown that these bacteria do not elicit an inflammatory response (H. Shin, M. Mally, M. Kuhn, C. Paroz, and G. R. Cornelis, J. Infect. Dis. 195:375-386, 2007). Here, we analyze their sensitivity to the innate immune system. Bacteria from the archetype strain Cc5 were highly resistant to killing by complement. There was little membrane attack complex (MAC) deposition in spite of C3b deposition. Cc5 bacteria were as resistant to phagocytosis by human polymorphonuclear leukocytes (PMNs) as Yersinia enterocolitica MRS40, endowed with an antiphagocytic type III secretion system. We isolated Y1C12, a transposon mutant that is hypersensitive to killing by complement via the antibody-dependent classical pathway. The mutation inactivated a putative glycosyltransferase gene, suggesting that the Y1C12 mutant was affected at the level of a capsular polysaccharide or lipopolysaccharide (LPS) structure. Cc5 appeared to have several polysaccharidic structures, one being altered in Y1C12. The structure missing in Y1C12 could be purified by classical LPS purification procedures and labeled by tritiated palmitate, indicating that it is more likely to be an LPS structure than a capsule. Y1C12 bacteria were also more sensitive to phagocytosis by PMNs than wild-type bacteria. In conclusion, a polysaccharide structure, likely an LPS, protects C. canimorsus from deposition of the complement MAC and from efficient phagocytosis by PMNs.


Subject(s)
Capnocytophaga/immunology , Complement System Proteins/immunology , Microbial Viability , Neutrophils/immunology , Animals , Bacterial Proteins/genetics , Blood Bactericidal Activity , Colony Count, Microbial , DNA Transposable Elements , Dogs , Glycosyltransferases/genetics , Humans , Lipopolysaccharides/isolation & purification , Lipopolysaccharides/metabolism , Molecular Sequence Data , Mutagenesis, Insertional
9.
Microbes Infect ; 11(4): 509-14, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19285152

ABSTRACT

Capnocytophaga canimorsus is a Gram-negative commensal of dog's mouth causing severe human infections. A strain isolated from a human fatal infection was recently shown to have a sialidase, to inhibit the bactericidal activity of macrophages and to block the release of nitric oxide by LPS-stimulated macrophages. The present study aimed at determining the prevalence of C. canimorsus in dogs and the occurrence of these hypothetical virulence factors. C. canimorsus could be retrieved from the saliva of 61 dogs out of 106 sampled. Like in clinical isolates, all dog strains had a sialidase and 60% blocked the killing of phagocytosed Escherichia coli by macrophages. In contrast, only 6.5% of dog strains blocked the release of nitric oxide by LPS-challenged macrophages, suggesting that this property might contribute to virulence. The comparative analysis of 69 16S rDNA sequences revealed the existence of C. canimorsus strains that could be misdiagnosed.


Subject(s)
Capnocytophaga/isolation & purification , Dogs/microbiology , Saliva/microbiology , Virulence Factors/analysis , Animals , Bacterial Proteins/analysis , Capnocytophaga/genetics , Macrophages/immunology , Neuraminidase/analysis , Nitric Oxide/biosynthesis , Phagocytosis , Prevalence , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
10.
PLoS Pathog ; 4(9): e1000164, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18818736

ABSTRACT

Capnocytophaga canimorsus, a commensal bacterium of the canine oral flora, has been repeatedly isolated since 1976 from severe human infections transmitted by dog bites. Here, we show that C. canimorsus exhibits robust growth when it is in direct contact with mammalian cells, including phagocytes. This property was found to be dependent on a surface-exposed sialidase allowing C. canimorsus to utilize internal aminosugars of glycan chains from host cell glycoproteins. Although sialidase probably evolved to sustain commensalism, by releasing carbohydrates from mucosal surfaces, it also contributed to bacterial persistence in a murine infection model: the wild type, but not the sialidase-deficient mutant, grew and persisted, both when infected singly or in competition. This study reveals an example of pathogenic bacteria feeding on mammalian cells, including phagocytes by deglycosylation of host glycans, and it illustrates how the adaptation of a commensal to its ecological niche in the host, here the dog's oral cavity, contributes to being a potential pathogen.


Subject(s)
Capnocytophaga/metabolism , Epithelial Cells/microbiology , Phagocytes/microbiology , Animals , Capnocytophaga/enzymology , Capnocytophaga/isolation & purification , Cells, Cultured , Gram-Negative Bacterial Infections , Humans , Mice , Neuraminidase/metabolism , Polysaccharides/metabolism
11.
Appl Environ Microbiol ; 74(20): 6369-77, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18723653

ABSTRACT

Capnocytophaga canimorsus, a commensal bacterium from canine oral flora, has been isolated throughout the world from severe human infections caused by dog bites. Due to the low level of evolutionary relationship to Proteobacteria, genetic methods suitable for the genus Capnocytophaga needed to be established. Here, we show that Tn4351, derived from Bacteroides fragilis, could be introduced by conjugation into C. canimorsus and conferred resistance to erythromycin. By mapping and sequencing a naturally occurring plasmid isolated from a clinical isolate of C. canimorsus, we identified a repA gene that allowed us to construct Escherichia coli-Capnocytophaga shuttle vectors. Most commonly used antibiotic markers were not functional in C. canimorsus, but cefoxitin (cfxA), tetracycline (tetQ), and erythromycin (ermF) resistances could be used as markers for plasmid maintenance in C. canimorsus and even in some other Capnocytophaga spp. Shuttle vectors were introduced into C. canimorsus either by conjugation using the origin of transfer (oriT) of RP4 or by electrotransformation. Taking advantage of the promoter of ermF, an expression vector was constructed. Finally, a method that allows site-directed mutagenesis is described. All these genetic tools pave the way, not only for molecular studies of the pathogenesis of C. canimorsus, but also for studies of other oral Capnocytophaga species.


Subject(s)
Capnocytophaga/genetics , Molecular Biology/methods , Animals , Anti-Bacterial Agents/pharmacology , Base Sequence , Capnocytophaga/isolation & purification , Conjugation, Genetic , DNA Transposable Elements , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Dogs , Drug Resistance, Bacterial , Erythromycin/pharmacology , Escherichia coli/genetics , Gene Expression , Genetic Vectors , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Plasmids , Selection, Genetic , Sequence Analysis, DNA
12.
J Infect Dis ; 195(3): 375-86, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17205476

ABSTRACT

Capnocytophaga canimorsus, a commensal bacterium from dogs' mouths, can cause septicemia or meningitis in humans through bites or scratches. Here, we describe and characterize the inflammatory response of human and mouse macrophages on C. canimorsus infection. Macrophages infected with 10 different strains failed to release tumor necrosis factor (TNF)- alpha and interleukin (IL)-1 alpha . Macrophages infected with live and heat-killed (HK) C. canimorsus 5 (Cc5), a strain isolated from a patient with fatal septicemia, did not release IL-6, IL-8, interferon- gamma , macrophage inflammatory protein-1 beta , and nitric oxide (NO). This absence of a proinflammatory response was characterized by the inability of Toll-like receptor (TLR) 4 to respond to Cc5. Moreover, live but not HK Cc5 blocked the release of TNF- alpha and NO induced by HK Yersinia enterocolitica. In addition, live Cc5 down-regulated the expression of TLR4 and dephosphorylated p38 mitogen-activated protein kinase. These results highlight passive and active mechanisms of immune evasion by C. canimorsus, which may explain its capacity to escape from the host immune system.


Subject(s)
Capnocytophaga , Gram-Negative Bacterial Infections/immunology , Animals , Capnocytophaga/pathogenicity , Cells, Cultured , Chemokine CCL4 , Dogs , Down-Regulation , Gram-Negative Bacterial Infections/metabolism , Humans , Interferon-gamma/biosynthesis , Interleukin-1alpha/biosynthesis , Interleukin-8/biosynthesis , Macrophage Inflammatory Proteins/biosynthesis , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Nitric Oxide/biosynthesis , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Virulence , p38 Mitogen-Activated Protein Kinases/metabolism
13.
J Bacteriol ; 188(17): 6346-53, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16923902

ABSTRACT

Bacteria commonly exchange genetic information by the horizontal transfer of conjugative plasmids. In gram-negative conjugation, a relaxase enzyme is absolutely required to prepare plasmid DNA for transit into the recipient via a type IV secretion system. Here we report a mutagenesis of the F plasmid relaxase gene traI using in-frame, 31-codon insertions. Phenotypic analysis of our mutant library revealed that several mutant proteins are functional in conjugation, highlighting regions of TraI that can tolerate insertions of a moderate size. We also demonstrate that wild-type TraI, when overexpressed, plays a dominant-negative regulatory role in conjugation, repressing plasmid transfer frequencies approximately 100-fold. Mutant TraI proteins with insertions in a region of approximately 400 residues between the consensus relaxase and helicase sequences did not cause conjugative repression. These unrestrictive TraI variants have normal relaxase activity in vivo, and several have wild-type conjugative functions when expressed at normal levels. We postulate that TraI negatively regulates conjugation by interacting with and sequestering some component of the conjugative apparatus. Our data indicate that the domain responsible for conjugative repression resides in the central region of TraI between the protein's catalytic domains.


Subject(s)
DNA Helicases/genetics , Escherichia coli/genetics , F Factor/genetics , Conjugation, Genetic/physiology , DNA Helicases/physiology , Down-Regulation , Escherichia coli Proteins , Mutagenesis
SELECTION OF CITATIONS
SEARCH DETAIL
...