Network


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

Hotspot


Dive into the research topics where A.J.M. Ligtenberg is active.

Publication


Featured researches published by A.J.M. Ligtenberg.


Biological Chemistry | 2007

Salivary agglutinin/glycoprotein-340/DMBT1: a single molecule with variable composition and with different functions in infection, inflammation and cancer.

A.J.M. Ligtenberg; Enno C. I. Veerman; Arie V. Nieuw Amerongen; Jan Mollenhauer

Abstract Salivary agglutinin (SAG), lung glycoprotein-340 (gp-340) and Deleted in Malignant Brain Tumours 1 (DMBT1) are three names for identical proteins encoded by the dmbt1 gene. DMBT1/SAG/gp-340 belongs to the scavenger receptor cysteine-rich (SRCR) superfamily of proteins, a superfamily of secreted or membrane-bound proteins with SRCR domains that are highly conserved down to sponges, the most ancient metazoa. On the one hand, DMBT1 may represent an innate defence factor acting as a pattern recognition molecule. It interacts with a broad range of pathogens, including cariogenic streptococci and Helicobacter pylori, influenza viruses and HIV, but also with mucosal defence proteins, such as IgA, surfactant proteins and MUC5B. Stimulation of alveolar macrophage migration, suppression of neutrophil oxidative burst and activation of the complement cascade point further to an important role in the regulation of inflammatory responses. On the other hand, DMBT1 has been demonstrated to play a role in epithelial and stem cell differentiation. Inactivation of the gene coding for this protein may lead to disturbed differentiation, possibly resulting in tumour formation. These data strongly point to a role for DMBT1 as a molecule linking innate immune processes with regenerative processes.


Biochemical Journal | 2006

Salivary agglutinin and lung scavenger receptor cysteine-rich glycoprotein 340 have broad anti-influenza activities and interactions with surfactant protein D that vary according to donor source and sialylation.

Kevan L. Hartshorn; A.J.M. Ligtenberg; Mitchell R. White; Martin van Eijk; Max Hartshorn; Lily Pemberton; Uffe Holmskov; Erika C. Crouch

We previously found that scavenger receptor cysteine-rich gp-340 (glycoprotein-340), isolated from lung or saliva, directly inhibits human IAVs (influenza A viruses). We now show that salivary gp-340 has broad antiviral activity against human, equine and porcine IAV strains. Although lung and salivary gp-340 are identical in protein sequence, salivary gp-340 from one donor had significantly greater antiviral activity against avian-like IAV strains which preferentially bind sialic acids in alpha(2,3) linkage. A greater density of alpha(2,3)-linked sialic acids was present on the salivary gp-340 from this donor as compared with salivary gp-340 from another donor or several preparations of lung gp-340. Hence, the specificity of sialic acid linkages on gp-340 is an important determinant of anti-IAV activity. Gp-340 binds to SP-D (surfactant protein D), and we previously showed that lung gp-340 has co-operative interactions with SP-D in viral neutralization and aggregation assays. We now report that salivary gp-340 can, in some cases, strongly antagonize certain antiviral activities of SP-D. This effect was associated with greater binding of salivary gp-340 to the carbohydrate recognition domain of SP-D as compared with the binding of lung gp-340. These findings may relate to inter-individual variations in innate defence against highly pathogenic IAV and to effects of aspiration of oral contents on SP-D-mediated lung functions.


Journal of Dental Research | 2002

Immunohistochemical Detection of Salivary Agglutinin/gp-340 in Human Parotid, Submandibular, and Labial Salivary Glands

Floris J. Bikker; A.J.M. Ligtenberg; J.E. van der Wal; P.A.M. van den Keijbus; Uffe Holmskov; Enno C. I. Veerman; A.V. Nieuw Amerongen

Salivary agglutinin is a Streptococcus mutans binding protein and a member of the scavenger receptor cysteine-rich superfamily. It is identical to lung gp-340 and brain DMBT1, which possibly play a role in innate immunity and tumor suppression, respectively. The goal of this study was to localize salivary agglutinin in human salivary glands. Two monoclonal antibodies, directed against gp-340, were characterized. mAb 213-1 reacted with sialic acid epitopes and cross-reacted with MUC7. The reaction with mAb 213-6 disappeared after reduction, suggesting that a protein epitope was recognized. In the parotid gland, immunohistochemical labeling with mAb 213-6 was found in the duct cells. In the submandibular gland and labial gland, both serous acini and demilune cells were labeled. In the labial gland, labeling was found at the luminal side of the duct cells. Salivary agglutinin was distinctly localized in salivary glands, but in distinct glandular secretions, no differences in electrophoretic behavior were observed.


Antonie Van Leeuwenhoek International Journal of General and Molecular Microbiology | 1996

Interaction of the salivary low-molecular-weight mucin (MG2) with Actinobacillus actinomycetemcomitans

Jasper Groenink; A.J.M. Ligtenberg; Enno C. I. Veerman; J.G.M. Bolscher; A.V. Nieuw Amerongen

Periodontitis is associated with the presence of certain Gram-negative bacteria in the oral cavity, among these Actinobacillus actinomycetemcomitans. In order to determine which types of salivary components interact with A. actinomycetemcomitans two strains (HG 1175 and FDC Y4) were incubated with whole saliva and individual glandular secretions, viz. parotid, submandibular, and sublingual saliva. Immunochemical analysis by immunoblotting of bacteria-bound salivary proteins showed that IgA, the low-molecular mucin MG2, parotid agglutinin, and a 300 kDa sublingual and submandibular glycoprotein, were bound to the bacterial strains tested. In addition, adherence of A. actinomycetemcomitans to salivary proteins in a solid-phase was studied. After electrophoresis and transfer of salivary proteins to nitrocellulose membranes A. actinomycetemcomitans adhered only to MG2. In this assay periodate treatment, mild acid hydrolysis or neuraminidase digestion of the saliva glycoproteins abolished binding of two clinical isolates (HG 1175 and NY 664), suggesting that sialic acid residues on MG2 are involved in the binding. In contrast, adherence of the smooth laboratory strain Y4 was not affected by removal of sialic acid residues or even periodate treatment of MG2.


European Journal of Immunology | 2009

DMBT1 functions as pattern-recognition molecule for poly-sulfated and poly-phosphorylated ligands.

Caroline End; Floris J. Bikker; Marcus Renner; Gaby Bergmann; Stefan Lyer; Stephanie Blaich; Melanie Hudler; Burkhard Helmke; Nikolaus Gassler; Frank Autschbach; A.J.M. Ligtenberg; Axel Benner; Uffe Holmskov; Peter Schirmacher; Arie V. Nieuw Amerongen; Philip Rosenstiel; Christian Sina; Andre Franke; Mathias Hafner; Petra Kioschis; Stefan Schreiber; Annemarie Poustka; Jan Mollenhauer

Deleted in malignant brain tumors 1 (DMBT1) is a secreted glycoprotein displaying a broad bacterial‐binding spectrum. Recent functional and genetic studies linked DMBT1 to the suppression of LPS‐induced TLR4‐mediated NF‐κB activation and to the pathogenesis of Crohns disease. Here, we aimed at unraveling the molecular basis of its function in mucosal protection and of its broad pathogen‐binding specificity. We report that DMBT1 directly interacts with dextran sulfate sodium (DSS) and carrageenan, a structurally similar sulfated polysaccharide, which is used as a texturizer and thickener in human dietary products. However, binding of DMBT1 does not reduce the cytotoxic effects of these agents to intestinal epithelial cells in vitro. DSS and carrageenan compete for DMBT1‐mediated bacterial aggregation via interaction with its bacterial‐recognition motif. Competition and ELISA studies identify poly‐sulfated and poly‐phosphorylated structures as ligands for this recognition motif, such as heparansulfate, LPS, and lipoteichoic acid. Dose–response studies in Dmbt1−/− and Dmbt1+/+ mice utilizing the DSS‐induced colitis model demonstrate a differential response only to low but not to high DSS doses. We propose that DMBT1 functions as pattern‐recognition molecule for poly‐sulfated and poly‐phosphorylated ligands providing a molecular basis for its broad bacterial‐binding specificity and its inhibitory effects on LPS‐induced TLR4‐mediated NF‐κB activation.


International Journal of Molecular Sciences | 2010

Deleted in Malignant Brain Tumors-1 Protein (DMBT1): A Pattern Recognition Receptor with Multiple Binding Sites

A.J.M. Ligtenberg; Niclas G. Karlsson; Enno C. I. Veerman

Deleted in Malignant Brain Tumors-1 protein (DMBT1), salivary agglutinin (DMBT1SAG), and lung glycoprotein-340 (DMBT1GP340) are three names for glycoproteins encoded by the same DMBT1 gene. All these proteins belong to the scavenger receptor cysteine-rich (SRCR) superfamily of proteins: a superfamily of secreted or membrane-bound proteins with SRCR domains that are highly conserved down to sponges, the most ancient metazoa. In addition to SRCR domains, all DMBT1s contain two CUB domains and one zona pellucida domain. The SRCR domains play a role in the function of DMBT1s, which is the binding of a broad range of pathogens including cariogenic streptococci, Helicobacter pylori and HIV. Mucosal defense proteins like IgA, surfactant proteins and lactoferrin also bind to DMBT1s through their SRCR domains. The binding motif on the SRCR domains comprises an 11-mer peptide in which a few amino acids are essential for binding (GRVEVLYRGSW). Adjacent to each individual SRCR domain are glycosylation domains, where the attached carbohydrate chains play a role in the binding of influenza A virus and Helicobacter pylori. The composition of the carbohydrate chains is not only donor specific, but also varies between different organs. These data demonstrate a role for DMBT1s as pattern recognition molecules containing various peptide and carbohydrate binding motifs.


Annals of the New York Academy of Sciences | 2007

Implications for Diagnostics in the Biochemistry and Physiology of Saliva

Arie V. Nieuw Amerongen; A.J.M. Ligtenberg; Enno C. I. Veerman

Abstract:  Oral fluid mainly consists of a mixture of glandular salivas. In addition, it is contaminated by some crevicular fluid, containing serum constituents. The contribution of the various salivary glands shows a continuous variation, resulting in wide ranges of concentrations for all constituents of oral fluid. As a consequence, the collection of oral fluid for diagnostic purposes should be standardized. Oral fluid can be used to detect a number of diseases and recent use of illicit drugs. It can also be used to monitor therapeutic drug concentrations. The development of microchips for salivary components offers great possibilities to use oral fluid for point‐of‐care testing.


Genes, Chromosomes and Cancer | 2004

Carcinogen inducibility in vivo and down-regulation of DMBT1 during breast carcinogenesis

Jan Mollenhauer; Burkhard Helmke; Daniel Medina; Gaby Bergmann; Nikolaus Gassler; Hanna Müller; Stefan Lyer; Laura Diedrichs; Marcus Renner; Rainer Wittig; Stephanie Blaich; Ute Hamann; Jens Madsen; Uffe Holmskov; Floris J. Bikker; A.J.M. Ligtenberg; Anette Carlén; Jan Olsson; Herwart F. Otto; Bert W. O'Malley; Annemarie Poustka

Deleted in malignant brain tumors 1 (DMBT1) has been proposed as a candidate tumor suppressor for brain and epithelial cancer. Initial studies suggested loss of expression rather than mutation as the predominant mode of DMBT1 inactivation. However, in situ studies in lung cancer demonstrated highly sophisticated changes of DMBT1 expression and localization, pointing to a chronological order of events. Here we report on the investigation of DMBT1 in breast cancer in order to test whether these principles might also be attributable to other tumor types. Comprehensive mutational analyses did not uncover unambiguous inactivating DMBT1 mutations in breast cancer. Expression analyses in the human and mouse mammary glands pointed to the necessity of DMBT1 induction. While age‐dependent and hormonal effects could be ruled out, 9 of 10 mice showed induction of Dmbt1 expression after administration of the carcinogen 7,12‐dimethybenz(α)anthracene prior to the onset of tumorigenesis or other histopathological changes. DMBT1 displayed significant up‐regulation in human tumor–flanking tissues compared to in normal breast tissues (P < 0.05). However, the breast tumor cells displayed a switch from lumenal secretion to secretion to the extracellular matrix and a significant down‐regulation compared to that in matched normal flanking tissues (P < 0.01). We concluded that loss of expression also is the predominant mode of DMBT1 inactivation in breast cancer. The dynamic behavior of DMBT1 in lung carcinoma is fully reflected in breast cancer, which suggests that this behavior might be common to tumor types arising from monolayered epithelia.


Biological Chemistry | 2008

A common binding motif for various bacteria of the bacteria-binding peptide SRCRP2 of DMBT1/gp-340/salivary agglutinin

J.T.D. Leito; A.J.M. Ligtenberg; Kamran Nazmi; J.M.A. de Blieck-Hogervorst; Enno C. I. Veerman; A. van Nieuw Amerongen

Abstract Salivary agglutinin (DMBT1SAG) is identical to lung glycoprotein-340 and encoded by the deleted in malignant brain tumors-1 gene. It is a member of the scavenger receptor cysteine-rich (SRCR) superfamily, proteins that have one or more SRCR domains. Salivary agglutinin plays a role in oral innate immunity by the binding and agglutination of oral streptococci. Streptococcus mutans has been shown to bind to a 16-mer peptide (QGRVEVLYRGSWGTVC) located within the SRCR domains. Within this peptide, designated SRCR peptide 2, residues VEVL and W are critical for binding. The aim of this study was to investigate binding of DMBT1SAG to other bacteria. Therefore, interaction between a series of bacteria and DMBT1SAG, SRCR peptide 2 and its alanine substitution variants was investigated in adhesion and agglutination assays. For different bacteria there was a highly significant correlation between adhesion to DMBT1SAG and adhesion to SRCR peptide 2, suggesting that SRCR peptide 2 is the major bacteria-binding site. An alanine substitution scan showed that eight amino acids are involved in binding (xRVEVLYxxSWxxxx). The binding motifs varied for different species, but the residues VxVxY and W are always present. In conclusion, a common binding motif (RVEVLYxxxSW) within the SRCR domains is responsible for the broad bacteria-binding spectrum of DMBT1SAG.


Biochemical Journal | 2004

Binding of salivary agglutinin to IgA

A.J.M. Ligtenberg; Floris J. Bikker; Jolanda M.A. de Blieck-Hogervorst; Enno C. I. Veerman; Arie V. Nieuw Amerongen

SAG (salivary agglutinin), which is identical to gp-340 (glycoprotein-340) from the lung, is encoded by DMBT1 (deleted in malignant brain tumours 1). It is a member of the SRCR (scavenger receptor cysteine-rich) superfamily and contains 14 SRCR domains, 13 of which are highly similar. SAG in saliva is partially complexed with IgA, which may be necessary for bacterial binding. The goal of the present study was to characterize the binding of purified SAG to IgA. SAG binds to a variety of proteins, including serum and secretory IgA, alkaline phosphatase-conjugated IgGs originating from rabbit, goat, swine and mouse, and lactoferrin and albumin. Binding of IgA to SAG is calcium dependent and is inhibited by 0.5 M KCl, suggesting that electrostatic interactions are involved. Binding of IgA was destroyed after reduction of SAG, suggesting that the protein moiety is involved in binding. To pinpoint further the binding domain for IgA on SAG, a number of consensus-based peptides of the SRCR domains and SRCR interspersed domains were designed and synthesized. ELISA binding studies with IgA indicated that only one of the peptides tested, comprising amino acids 18-33 (QGRVEVLYRGSWGTVC) of the 109-amino-acid SRCR domain, exhibited binding to IgA. This domain is identical to the domain of SAG that is involved in binding to bacteria. Despite this similar binding site, IgA did not inhibit binding of Streptococcus mutans to SAG or peptide. These results show that the binding of IgA to SAG is specifically mediated by a peptide sequence on the SRCR domains.

Collaboration


Dive into the A.J.M. Ligtenberg's collaboration.

Top Co-Authors

Avatar

Enno C. I. Veerman

Academic Center for Dentistry Amsterdam

View shared research outputs
Top Co-Authors

Avatar

Floris J. Bikker

Academic Center for Dentistry Amsterdam

View shared research outputs
Top Co-Authors

Avatar

Jan Mollenhauer

University of Southern Denmark

View shared research outputs
Top Co-Authors

Avatar

Kamran Nazmi

Academic Center for Dentistry Amsterdam

View shared research outputs
Top Co-Authors

Avatar

Annemarie Poustka

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Stefan Lyer

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Uffe Holmskov

University of Southern Denmark

View shared research outputs
Top Co-Authors

Avatar

A.V. Nieuw Amerongen

Academic Center for Dentistry Amsterdam

View shared research outputs
Top Co-Authors

Avatar

Arie V. Nieuw Amerongen

Academic Center for Dentistry Amsterdam

View shared research outputs
Top Co-Authors

Avatar

Marcus Renner

University Hospital Heidelberg

View shared research outputs
Researchain Logo
Decentralizing Knowledge