Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Michael Lebens is active.

Publication


Featured researches published by Michael Lebens.


Nature | 2011

Evidence for several waves of global transmission in the seventh cholera pandemic

Ankur Mutreja; Dong Wook Kim; Nicholas R. Thomson; Thomas Richard Connor; Je Hee Lee; Samuel Kariuki; Nicholas J. Croucher; Seon Young Choi; Simon R. Harris; Michael Lebens; Swapan Kumar Niyogi; Eun Jin Kim; Thandavarayan Ramamurthy; Jongsik Chun; J. L. N. Wood; John D. Clemens; Cecil Czerkinsky; G. Balakrish Nair; Jan Holmgren; Julian Parkhill; Gordon Dougan

Vibrio cholerae is a globally important pathogen that is endemic in many areas of the world and causes 3–5 million reported cases of cholera every year. Historically, there have been seven acknowledged cholera pandemics; recent outbreaks in Zimbabwe and Haiti are included in the seventh and ongoing pandemic. Only isolates in serogroup O1 (consisting of two biotypes known as ‘classical’ and ‘El Tor’) and the derivative O139 (refs 2, 3) can cause epidemic cholera. It is believed that the first six cholera pandemics were caused by the classical biotype, but El Tor has subsequently spread globally and replaced the classical biotype in the current pandemic. Detailed molecular epidemiological mapping of cholera has been compromised by a reliance on sub-genomic regions such as mobile elements to infer relationships, making El Tor isolates associated with the seventh pandemic seem superficially diverse. To understand the underlying phylogeny of the lineage responsible for the current pandemic, we identified high-resolution markers (single nucleotide polymorphisms; SNPs) in 154 whole-genome sequences of globally and temporally representative V. cholerae isolates. Using this phylogeny, we show here that the seventh pandemic has spread from the Bay of Bengal in at least three independent but overlapping waves with a common ancestor in the 1950s, and identify several transcontinental transmission events. Additionally, we show how the acquisition of the SXT family of antibiotic resistance elements has shaped pandemic spread, and show that this family was first acquired at least ten years before its discovery in V. cholerae.


Infection and Immunity | 2001

Cholera Toxin B Subunit as a Carrier Molecule Promotes Antigen Presentation and Increases CD40 and CD86 Expression on Antigen-Presenting Cells

Annie George-Chandy; Kristina Eriksson; Michael Lebens; Inger Nordström; Emma Schön; Jan Holmgren

ABSTRACT Cholera toxin B subunit (CTB) is an efficient mucosal carrier molecule for the generation of mucosal antibody responses and/or induction of systemic T-cell tolerance to linked antigens. CTB binds with high affinity to GM1 ganglioside cell surface receptors. In this study, we evaluated how conjugation of a peptide or protein antigen to CTB by chemical coupling or genetic fusion influences the T-cell-activating capacity of different antigen-presenting cell (APC) subsets. Using an in vitro system in which antigen-pulsed APCs were incubated with antigen-specific, T-cell receptor-transgenic T cells, we found that the dose of antigen required for T-cell activation could be decreased >10,000-fold using CTB-conjugated compared to free antigen. In contrast, no beneficial effects were observed when CTB was simply admixed with antigen. CTB conjugation enhanced the antigen-presenting capacity not only of dendritic cells and B cells but also of macrophages, which expressed low levels of cell surface major histocompatibility complex (MHC) class II and were normally poor activators of naive T cells. Enhanced antigen-presenting activity by CTB-linked antigen resulted in both increased T-cell proliferation and increased interleukin-12 and gamma interferon secretion and was associated with up-regulation of CD40 and CD86 on the APC surface. These results imply that conjugation to CTB dramatically lowers the threshold concentration of antigen required for immune cell activation and also permits low-MHC II-expressing APCs to prime for a specific immune response.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2010

Intranasal Immunization With an Apolipoprotein B-100 Fusion Protein Induces Antigen-Specific Regulatory T Cells and Reduces Atherosclerosis

R Klingenberg; Michael Lebens; Andreas Hermansson; Gunilla Nordin Fredrikson; Daniela Strodthoff; Mats Rudling; Daniel F.J. Ketelhuth; Jan Holmgren; Jan Nilsson; Göran K. Hansson

Objective—Atherosclerosis is an inflammatory disease. Autoimmune responses to low-density lipoproteins (LDL) contribute to its progression, whereas immunization with LDL may induce atheroprotective or proatherogenic responses. The objective of this study was to develop an atheroprotective vaccine by targeting a peptide of the LDL protein constituent apolipoprotein B-100 (apoB-100) to the nasal mucosa to induce a protective mucosal immune response. Methods and Results—A peptide comprising amino acids 3136 to 3155 of apoB-100 (p210) was fused to the B subunit of cholera toxin (CTB), which binds to a ganglioside on mucosal epithelia. The effect of nasal administration of the p210-CTB fusion protein on atherogenesis was compared with that of an ovalbumin peptide fused to CTB and with untreated controls. Immunization with p210-CTB for 12 weeks caused a 35% reduction in aortic lesion size in Apoe−/− mice. This effect was accompanied by induction of regulatory T cells that markedly suppressed effector T cells rechallenged with apoB-100 and increased numbers of interleukin (IL)-10+ CD4+ T cells. Furthermore, a peptide-specific antibody response was observed. Atheroprotection was also documented in apoe−/− mice lacking functional transforming growth factor-&bgr; receptors on T cells. Conclusion—Nasal administration of an apoB-100 peptide fused to CTB attenuates atherosclerosis and induces regulatory Tr1 cells that inhibit T effector responses to apoB-100.


Journal of Immunology | 2004

In Vivo Adjuvant-Induced Mobilization and Maturation of Gut Dendritic Cells after Oral Administration of Cholera Toxin

Fabienne Anjuère; Carmelo Luci; Michael Lebens; Déborah Rousseau; Catherine Hervouet; Geneviève Milon; Jan Holmgren; Carlos Ardavín; Cecil Czerkinsky

Although dendritic cells (DCs) regulate immune responses, they exhibit functional heterogeneity depending on their anatomical location. We examined the functional properties of intestinal DCs after oral administration of cholera toxin (CT), the most potent mucosal adjuvant. Two CD11c+ DC subsets were identified both in Peyer’s patches and mesenteric lymph nodes (MLN) based on the expression of CD8α (CD8+ and CD8− DCs, respectively). A third subset of CD11c+CD8int was found exclusively in MLN. Feeding mice with CT induced a rapid and transient mobilization of a new CD11c+CD8− DC subset near the intestinal epithelium. This recruitment was associated with an increased production of the chemokine CCL20 in the small intestine and was followed by a massive accumulation of CD8int DCs in MLN. MLN DCs from CT-treated mice were more potent activators of naive T cells than DCs from control mice and induced a Th2 response. This increase in immunostimulating properties was accounted for by CD8int and CD8− DCs, whereas CD8+ DCs remained insensitive to CT treatment. Consistently, the CD8int and CD8− subsets expressed higher levels of costimulatory molecules than CD8+ and corresponding control DCs. Adoptive transfer experiments showed that these two DC subsets, unlike CD8+ DCs, were able to present Ags orally coadministered with CT in an immunostimulating manner. The ability of CT to mobilize immature DCs in the intestinal epithelium and to promote their emigration and differentiation in draining lymph nodes may explain the exceptional adjuvant properties of this toxin on mucosal immune responses.


Infection and Immunity | 2006

The Major Subunit, CfaB, of Colonization Factor Antigen I from Enterotoxigenic Escherichia coli Is a Glycosphingolipid Binding Protein

Lena Jansson; Joshua Tobias; Michael Lebens; Ann-Mari Svennerholm; Susann Teneberg

ABSTRACT Bacterial adherence to mucosal surfaces is an important virulence trait of pathogenic bacteria. Adhesion of enterotoxigenic Escherichia coli (ETEC) to the intestine is mediated by a number of antigenically distinct colonization factors (CFs). One of the most common CFs is CFA/I. This has a fimbrial structure composed of a major repeating subunit, CfaB, and a single tip subunit, CfaE. The potential carbohydrate recognition by CFA/I was investigated by binding CFA/I-fimbriated bacteria and purified CFA/I fimbriae to a large number of variant glycosphingolipids separated on thin-layer chromatograms. For both fimbriated bacteria and purified fimbriae, specific interactions could be identified with a number of nonacid glycosphingolipids. These included glucosylceramide, lactosylceramide with phytosphingosine and/or hydroxy fatty acids, neolactotetraosylceramide, gangliotriaosylceramide, gangliotetraosylceramide, the H5 type 2 pentaglycosylceramide, the Lea-5 glycosphingolipid, the Lex-5 glycosphingolipid, and the Ley-6 glycosphingolipid. These glycosphingolipids were also recognized by recombinant E. coli expressing CFA/I in the absence of tip protein CfaE, as well as by purified fimbriae from the same strain. This demonstrates that the glycosphingolipid-binding capacity of CFA/I resides in the major CfaB subunit.


Gene | 1994

Insertion of a HIV-1-neutralizing epitope in a surface-exposed internal region of the cholera toxin B-subunit.

Malin Bäckström; Michael Lebens; Florian Schödel; Jan Holmgren

The non-toxic B-subunit of cholera toxin (CTB) is a powerful immunogen and has been investigated as a carrier for foreign peptide epitopes, with peptides genetically fused to either the N- or C terminus of CTB. In the present study, we have constructed a plasmid encoding a novel intrachain CTB fusion protein with a peptide epitope inserted into an internal region of CTB: eight amino acids (aa) in CTB (56-63) were substituted with a 10-aa peptide from the third variable (V3) loop of the HIV-1 envelope protein gp120. The resulting chimeric protein retained important functional characteristics of the native CTB including pentamerization and GM1 ganglioside receptor binding. The internal hybrid protein was also shown to be resistant to proteolytic degradation during production in Vibrio cholerae, whereas a terminal hybrid protein, where the same gp120-epitope was fused to the N terminus of CTB, was rapidly cleaved during culture. The inserted epitope, which is known to give rise to HIV-1 neutralizing Ab, could be detected with a V3 loop-specific monoclonal Ab when the chimeric protein was analyzed in ELISA and immunoblot, indicating that the epitope inserted at this site is presented on the surface of the protein. Consistent with these observations, immunization of mice with the CTB::HIV hybrid protein elicited a high titered serum Ab response to the CTB moiety and also, in some but not all animals, a detectable response to the inserted gp120 epitope.


PLOS ONE | 2009

Sulfatide recognition by colonization factor antigen CS6 from enterotoxigenic Escherichia coli.

Lena Jansson; Joshua Tobias; Catharina Jarefjäll; Michael Lebens; Ann-Mari Svennerholm; Susann Teneberg

The first step in the pathogenesis of enterotoxigenic Escherichia coli (ETEC) infections is adhesion of the bacterium to the small intestinal epithelium. Adhesion of ETEC is mediated by a number of antigenically distinct colonization factors, and among these, one of the most commonly detected is the non-fimbrial adhesin coli surface antigen 6 (CS6). The potential carbohydrate recognition by CS6 was investigated by binding of recombinant CS6-expressing E. coli and purified CS6 protein to a large number of variant glycosphingolipids separated on thin-layer chromatograms. Thereby, a highly specific binding of the CS6-expressing E. coli, and the purified CS6 protein, to sulfatide (SO3-3Galβ1Cer) was obtained. The binding of the CS6 protein and CS6-expressing bacteria to sulfatide was inhibited by dextran sulfate, but not by dextran, heparin, galactose 4-sulfate or galactose 6-sulfate. When using recombinantly expressed and purified CssA and CssB subunits of the CS6 complex, sulfatide binding was obtained with the CssB subunit, demonstrating that the glycosphingolipid binding capacity of CS6 resides within this subunit. CS6-binding sulfatide was present in the small intestine of species susceptible to CS6-mediated infection, e.g. humans and rabbits, but lacking in species not affected by CS6 ETEC, e.g. mice. The ability of CS6-expressing ETEC to adhere to sulfatide in target small intestinal epithelium may thus contribute to virulence.


Vaccine | 2008

Construction of non-toxic Escherichia coli and Vibrio cholerae strains expressing high and immunogenic levels of enterotoxigenic E. coli colonization factor I fimbriae

Joshua Tobias; Michael Lebens; Gudrun Wiklund; Ann-Mari Svennerholm

To express high quantities of colonization factor antigen I (CFA/I) derived from enterotoxigenic Escherichia coli (ETEC) for use in ETEC vaccines, the entire CFA/I operon consisting of four genes (cfa-A, -B, -C, -E) was cloned into plasmid expression vectors that could be maintained either with or without antibiotic selection. Expression from the powerful tac promoter was under the control of the lacIq repressor present on the plasmids. Fimbriae were expressed on the surface of both a non-toxigenic E. coli K12 strain and a non-toxigenic strain of Vibrio cholerae following induction with isopropyl-beta-D-thiogalactopyranoside (IPTG). It was found that the recombinant E. coli strains expressed up to 16-fold higher levels of CFA/I fimbriae compared to a reference strain which had previously been shown to be among the highest natural producers of the CFA/I fimbriae among tested wild type ETEC strains. Oral immunization with formalin-killed recombinant E. coli bacteria over-expressing CFA/I induced significantly higher serum IgA and IgG+M antibodies responses compared to the reference strain. Oral immunization with formalin-killed recombinant V. cholerae bacteria also induce strong CFA/I-specific serum IgA and IgG+M responses. We conclude that our constructs may be useful as candidate strains in an oral killed CF-ETEC vaccine.


Expert Review of Vaccines | 2004

Current status and future prospects for a vaccine against schistosomiasis

Michael Lebens; Jia-Bin Sun; Cecil Czerkinsky; Jan Holmgren

Schistosomiasis remains an intractable problem in many parts of the world. Whereas the schistosome parasites cause little in the way of disease, their eggs become trapped in tissues of the host and elicit powerful and potentially damaging immune responses that are responsible for the pathology. Despite nearly four decades of effort there is still no effective vaccine against schistosomiasis, although a single vaccine candidate is undergoing clinical trials at present. Animal models have revealed much about disease progression and pathology. However, problems remain in identifying appropriate protective antigens to elicit immune responses that will attack the parasite but will not cross-react with egg antigens and thus increase the chance of developing severe chronic disease in individuals that have already been infected. This review summarizes the life-cycle of the parasite, current knowledge of pathogenesis and acquired immunity based on animal studies and observations in humans and the status of efforts in the vaccine development field.


Immunology | 2002

Genetic fusion of human insulin B-chain to the B-subunit of cholera toxin enhances in vitro antigen presentation and induction of bystander suppression in vivo

Hamid Sadeghi; Søren Bregenholt; Dale Wegmann; Jacob S. Petersen; Jan Holmgren; Michael Lebens

The pentameric B‐subunit of cholera toxin (CTB) can be used as an efficient mucosal carrier of either immunogenic or tolerogenic T‐cell epitopes. In this study a series of fusions was constructed between the genes encoding CTB and the B‐chain of human insulin (InsB). The resulting fusion proteins were expressed in Escherichia coli and isolated as cytoplasmic inclusion bodies that were then dissolved and assembled in vitro. GM1 enzyme‐linked immunosorbent assay (ELISA), sodium dodecyl sulphate–polyacrylamide gel electrophoresis (SDS–PAGE) and Western blot analyses showed that the protein construct in which InsB was fused to the C‐terminus of a CTB monomer (CI) assembled into structures that both bound to the receptor GM1 ganglioside and reacted with monoclonal antibodies to CTB and insulin. Fusion of InsB to the N‐terminus of CTB resulted in protein that could not assemble into pentameric CTB. In vitro assays showed that the CI fusion protein was 300‐fold more potent than native insulin at inducing interleukin‐2 (IL‐2) production by an insulin‐specific T‐cell hybridoma. When administered orally, the CI fusion protein induced efficient immunological suppression of ovalbumin‐specific T‐cell responses in mice co‐immunized parenterally with insulin and ovalbumin. These results demonstrate the stability, GM1 receptor‐binding activity and antigenic authenticity of the CI fusion protein as well as its ability to elicit insulin‐specific T‐cell responses in vitro. In addition, we demonstrate that the CI fusion protein induces efficient immunosuppression after oral administration, raising the possibility of using such constructs in the treatment of type‐1 diabetes.

Collaboration


Dive into the Michael Lebens's collaboration.

Top Co-Authors

Avatar

Jan Holmgren

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joshua Tobias

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jia-Bin Sun

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Erik Nygren

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lena Jansson

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge