Network


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

Hotspot


Dive into the research topics where Andrea Verhagen is active.

Publication


Featured researches published by Andrea Verhagen.


Journal of Biological Chemistry | 2012

Cross-comparison of protein recognition of sialic acid diversity on two novel sialoglycan microarrays.

Vered Padler-Karavani; Xuezheng Song; Hai Yu; Nancy Hurtado-Ziola; Shengshu Huang; Saddam Muthana; Harshal A. Chokhawala; Jiansong Cheng; Andrea Verhagen; Martijn A. Langereis; Ralf Kleene; Melitta Schachner; Raoul J. de Groot; Yi Lasanajak; Haruo Matsuda; Richard Schwab; Xi Chen; David F. Smith; Richard D. Cummings; Ajit Varki

Background: Various glycan microarrays are currently widely used, but systematic cross-comparisons are lacking. Results: We compare and contrast two sialoglycan microarrays using a variety of sialic acid-binding proteins. Conclusion: Diverse array formats can strengthen the quality of information, but differences between arrays may be observed. Significance: Glycan arrays with similar glycan structures cannot be simply assumed to give similar results. DNA and protein arrays are commonly accepted as powerful exploratory tools in research. This has mainly been achieved by the establishment of proper guidelines for quality control, allowing cross-comparison between different array platforms. As a natural extension, glycan microarrays were subsequently developed, and recent advances using such arrays have greatly enhanced our understanding of protein-glycan recognition in nature. However, although it is assumed that biologically significant protein-glycan binding is robustly detected by glycan microarrays, there are wide variations in the methods used to produce, present, couple, and detect glycans, and systematic cross-comparisons are lacking. We address these issues by comparing two arrays that together represent the marked diversity of sialic acid modifications, linkages, and underlying glycans in nature, including some identical motifs. We compare and contrast binding interactions with various known and novel plant, vertebrate, and viral sialic acid-recognizing proteins and present a technical advance for assessing specificity using mild periodate oxidation of the sialic acid chain. These data demonstrate both the diversity of sialic acids and the analytical power of glycan arrays, showing that different presentations in different formats provide useful and complementary interpretations of glycan-binding protein specificity. They also highlight important challenges and questions for the future of glycan array technology and suggest that glycan arrays with similar glycan structures cannot be simply assumed to give similar results.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Engagement of myelomonocytic Siglecs by tumor-associated ligands modulates the innate immune response to cancer

Heinz Läubli; Oliver M. T. Pearce; Flavio Schwarz; Shoib S. Siddiqui; Lingquan Deng; Michal A. Stanczak; Liwen Deng; Andrea Verhagen; Patrick Secrest; Chrissy Lusk; Ann G. Schwartz; Nissi M. Varki; Jack D. Bui; Ajit Varki

Significance In vitro and in vivo data indicate that hypersialylated tumor cells can engage Siglec-9 on myelomonocytic cells and influence the outcome of the interaction, depending on the stage of tumor growth and the microenvironment. On one hand, engagement of Siglec-9 or Siglec-E by tumor-associated ligands inhibited immunosurveillance and tumor cell killing during establishment of autologous tumors and new metastatic foci. On the other hand, inhibition of tumor-associated macrophages through Siglec-9 led to M1 polarization and reduced growth-promoting inflammation within the tumor microenvironment. This demonstrates a previously unidentified dualistic function of Siglec-9 during cancer progression. A functional polymorphism of Siglec-9 correlated with altered survival of lung cancer patients, suggesting that Siglec-9 might be therapeutically targeted. Certain pathogenic bacteria are known to modulate the innate immune response by decorating themselves with sialic acids, which can engage the myelomonocytic lineage inhibitory receptor Siglec-9, thereby evading immunosurveillance. We hypothesized that the well-known up-regulation of sialoglycoconjugates by tumors might similarly modulate interactions with innate immune cells. Supporting this hypothesis, Siglec-9–expressing myelomonocytic cells found in human tumor samples were accompanied by a strong up-regulation of Siglec-9 ligands. Blockade of Siglec-9 enhanced neutrophil activity against tumor cells in vitro. To investigate the function of inhibitory myelomonocytic Siglecs in vivo we studied mouse Siglec-E, the murine functional equivalent of Siglec-9. Siglec-E–deficient mice showed increased in vivo killing of tumor cells, and this effect was reversed by transgenic Siglec-9 expression in myelomonocytic cells. Siglec-E–deficient mice also showed enhanced immunosurveillance of autologous tumors. However, once tumors were established, they grew faster in Siglec-E–deficient mice. In keeping with this, Siglec-E–deficient macrophages showed a propensity toward a tumor-promoting M2 polarization, indicating a secondary role of CD33-related Siglecs in limiting cancer-promoting inflammation and tumor growth. Thus, we define a previously unidentified impact of inhibitory myelomonocytic Siglecs in cancer biology, with distinct roles that reflect the dual function of myelomonocytic cells in cancer progression. In keeping with this, a human polymorphism that reduced Siglec-9 binding to carcinomas was associated with improved early survival in non–small-cell lung cancer patients, which suggests that Siglec-9 might be therapeutically targeted within the right time frame and stage of disease.


The FASEB Journal | 2014

Rapid evolution of binding specificities and expression patterns of inhibitory CD33-related Siglecs in primates

Vered Padler-Karavani; Nancy Hurtado-Ziola; Yung-Chi Chang; Justin L. Sonnenburg; Arash Ronaghy; Hai Yu; Andrea Verhagen; Victor Nizet; Xi Chen; Nissi M. Varki; Ajit Varki; Takashi Angata

Siglecs are sialic acid‐binding Ig‐like lectins that recognize sialoglycans via amino‐terminal V‐set domains. CD33‐related Siglecs (CD33rSiglecs) on innate immune cells recognize endogenous sialoglycans as “self‐associated molecular patterns” (SAMPs), dampening immune responses via cytosolic immunoreceptor tyrosine‐based inhibition motifs that recruit tyrosine phosphatases. However, sialic acid‐expressing pathogens subvert this mechanism through molecular mimicry. Meanwhile, endogenous host SAMPs must continually evolve to evade other pathogens that exploit sialic acids as invasion targets. We hypothesized that these opposing selection forces have accelerated CD33rSiglec evolution. We address this by comparative analysis of major CD33rSiglec (Siglec‐3, Siglec‐5, and Siglec‐9) orthologs in humans, chimpanzees, and baboons. Recombinant soluble molecules displaying ligand‐binding domains show marked quantitative and qualitative interspecies differences in interactions with strains of the sialylated pathogen, group B Streptococcus, and with sialoglycans presented as gangliosides or in the form of sialoglycan microarrays, including variations such as N‐glycolyl and O‐acetyl groups. Primate Siglecs also show quantitative and qualitative intra‐ and interspecies variations in expression patterns on leukocytes, both in circulation and in tissues. Taken together our data explain why the CD33rSiglec‐encoding gene cluster is undergoing rapid evolution via multiple mechanisms, driven by the need to maintain self‐recognition by innate immune cells, while escaping 2 distinct mechanisms of pathogen subversion.—Padler‐Karavani, V., Hurtado‐Ziola, N., Chang, Y.‐C., Sonnenburg, J. L., Ronaghy, A., Yu, H., Verhagen, A., Nizet, V., Chen, X., Varki, N., Varki, A., Angata, T. Rapid evolution of binding specificities and expression patterns of inhibitory CD33‐related Siglecs in primates. FASEB J. 28, 1280–1293 (2014). www.fasebj.org


Proceedings of the National Academy of Sciences of the United States of America | 2012

Specific inactivation of two immunomodulatory SIGLEC genes during human evolution

Xiaoxia Wang; Nivedita Mitra; Ismael Secundino; Kalyan Banda; Pedro Cruz; Vered Padler-Karavani; Andrea Verhagen; Chris M. Reid; Martina Lari; Ermanno Rizzi; C. Balsamo; Giorgio Corti; Gianluca De Bellis; Laura Longo; William Beggs; David Caramelli; Sarah A. Tishkoff; Toshiyuki Hayakawa; Eric D. Green; James C. Mullikin; Victor Nizet; Jack D. Bui; Ajit Varki

Sialic acid-recognizing Ig-like lectins (Siglecs) are signaling receptors that modulate immune responses, and are targeted for interactions by certain pathogens. We describe two primate Siglecs that were rendered nonfunctional by single genetic events during hominin evolution after our common ancestor with the chimpanzee. SIGLEC13 was deleted by an Alu-mediated recombination event, and a single base pair deletion disrupted the ORF of SIGLEC17. Siglec-13 is expressed on chimpanzee monocytes, innate immune cells that react to bacteria. The human SIGLEC17P pseudogene mRNA is still expressed at high levels in human natural killer cells, which bridge innate and adaptive immune responses. As both resulting pseudogenes are homozygous in all human populations, we resurrected the originally encoded proteins and examined their functions. Chimpanzee Siglec-13 and the resurrected human Siglec-17 recruit a signaling adapter and bind sialic acids. Expression of either Siglec in innate immune cells alters inflammatory cytokine secretion in response to Toll-like receptor-4 stimulation. Both Siglecs can also be engaged by two potentially lethal sialylated bacterial pathogens of newborns and infants, agents with a potential impact on reproductive fitness. Neanderthal and Denisovan genomes show human-like sequences at both loci, corroborating estimates that the initial pseudogenization events occurred in the common ancestral population of these hominins. Both loci also show limited polymorphic diversity, suggesting selection forces predating the origin of modern humans. Taken together, these data suggest that genetic elimination of Siglec-13 and/or Siglec-17 represents signatures of infectious and/or other inflammatory selective processes contributing to population restrictions during hominin origins.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Inverse hormesis of cancer growth mediated by narrow ranges of tumor-directed antibodies

Oliver M. T. Pearce; Heinz Läubli; Andrea Verhagen; Patrick Secrest; Jiquan Zhang; Nissi M. Varki; Paul R. Crocker; Jack D. Bui; Ajit Varki

Significance We have previously shown that antibodies directed against tumor-associated antigens containing the nonhuman sialic acid N-glycolylneuraminic acid (Neu5Gc) can influence tumor progression. Here, we analyzed growth of Neu5Gc-positive tumors in Neu5Gc-deficient mice, following administration of increasing concentrations of anti-Neu5Gc antibodies. Although lower antibody concentrations stimulated tumor growth, higher concentrations inhibited growth, over a surprisingly narrow dose range. This biphasic narrow-range tumor growth response to tumor-directed antibodies (inverse hormesis) was reproduced in multiple mouse models, including one using a clinically approved monoclonal antibody. Our results are a novel experimental demonstration of how the levels of circulating antibodies can differentially influence tumor growth. These findings might have important implications in natural cancer progression and/or in cancer immunotherapy with antibodies. Compelling evidence for naturally occurring immunosurveillance against malignancies informs and justifies some current approaches toward cancer immunotherapy. However, some types of immune reactions have also been shown to facilitate tumor progression. For example, our previous studies showed that although experimental tumor growth is enhanced by low levels of circulating antibodies directed against the nonhuman sialic acid N-glycolyl-neuraminic acid (Neu5Gc), which accumulates in human tumors, growth could be inhibited by anti-Neu5Gc antibodies from a different source, in a different model. However, it remains generally unclear whether the immune responses that mediate cancer immunosurveillance vs. those responsible for inflammatory facilitation are qualitatively and/or quantitatively distinct. Here, we address this question using multiple murine tumor growth models in which polyclonal antibodies against tumor antigens, such as Neu5Gc, can alter tumor progression. We found that although growth was stimulated at low antibody doses, it was inhibited by high doses, over a linear and remarkably narrow range, defining an immune response curve (IRC; i.e., inverse hormesis). Moreover, modulation of immune responses against the tumor by altering antibody avidity or by enhancing innate immunity shifted the IRC in the appropriate direction. Thus, the dualistic role of immunosurveillance vs. inflammation in modulating tumor progression can be quantitatively distinguished in multiple model systems, and can occur over a remarkably narrow range. Similar findings were made in a human tumor xenograft model using a narrow range of doses of a monoclonal antibody currently in clinical use. These findings may have implications for the etiology, prevention, and treatment of cancer.


Analytical Chemistry | 2013

Quantum Dot Nanometal Surface Energy Transfer Based Biosensing of Sialic Acid Compositions and Linkages in Biological Samples

Raghavendra Kikkeri; Vered Padler-Karavani; Sandra Diaz; Andrea Verhagen; Hai Yu; Hongzhi Cao; Martijn A. Langereis; Raoul J. de Groot; Xi Chen; Ajit Varki

Current methods for analyzing sialic acid diversity in modifications and linkages require multistep processing, derivatization, and chromatographic analyses. We here report a single-step optical method for identification and quantification of different compositions of sialoglycans on glycoproteins and in serum. This was achieved by measuring and quantifying nanometal surface energy transfer (NSET) signals between quantum dots and gold nanoparticles bound to specific sialic acid binding proteins (SBPs) and sialic acid moieties, respectively. The biosensing process is based on the NSET turn-on by external sialic acid species that compete for binding to the SBPs. Selectivity of the biosensor toward sialoglycans can be designed to detect the total amount, glycosylation linkages (α2-6 vs α2-3), and modifications (9-O-acetyl and N-glycolyl groups) in the samples. This nanobiosensor is a prototype expected to achieve limits of the detection down to the micromolar range for high-throughput quantification and analysis of different compositions of sialoglycans present in biological or biomedical samples.


Journal of Biological Chemistry | 2014

Lectin Galactoside-binding Soluble 3 Binding Protein (LGALS3BP) Is a Tumor-associated Immunomodulatory Ligand for CD33-related Siglecs

Heinz Läubli; Frederico Alisson-Silva; Michal A. Stanczak; Shoib S. Siddiqui; Liwen Deng; Andrea Verhagen; Nissi M. Varki; Ajit Varki

Background: Engagement of inhibitory CD33-related Siglecs on immune cells has been shown to influence interactions with cancer cells, including tumor immune evasion. Results: LGALS3BP binds with high affinity to CD33-related Siglecs and inhibits neutrophil activation. Conclusion: We identify LGALS3BP as novel, cancer-associated Siglec ligand that can influence neutrophil activation. Significance: The engagement of inhibitory CD33-related Siglecs by LGALS3BP could support immune evasion of tumor cells. Lectin galactoside-binding soluble 3 binding protein (LGALS3BP, also called Mac-2 binding protein) is a heavily glycosylated secreted molecule that has been shown previously to be up-regulated in many cancers and has been implicated in tumor metastatic processes, as well as in other cell adhesion and immune functions. The CD33-related subset of sialic acid-binding immunoglobulin-like lectins (Siglecs) consists of immunomodulatory molecules that have recently been associated with the modulation of immune responses to cancer. Because up-regulation of Siglec ligands in cancer tissue has been observed, the characterization of these cancer-associated ligands that bind to inhibitory CD33-related Siglecs could provide novel targets for cancer immunomodulatory therapy. Here we used affinity chromatography of tumor cell extracts to identify LGALS3BP as a novel sialic acid-dependent ligand for human Siglec-9 and for other immunomodulatory Siglecs, such as Siglec-5 and Siglec-10. In contrast, the mouse homolog Siglec-E binds to murine LGALS3BP with lower affinity. LGALS3BP has been observed to be up-regulated in human colorectal and prostate cancer specimens, particularly in the extracellular matrix. Finally, LGALS3BP was able to inhibit neutrophil activation in a sialic acid- and Siglec-dependent manner. These findings suggest a novel immunoinhibitory function for LGALS3BP that might be important for immune evasion of tumor cells during cancer progression.


Methods in Enzymology | 2003

Recombinant influenza C hemagglutinin-esterase as a probe for sialic acid 9-O-acetylation

Laura Martin; Andrea Verhagen; Ajit Varki

Publisher Summary The binding and cleavage properties of the Influenza C virions hemagglutinin- esterase (CHE) protein for 9-O-acetyl groups on sialic acids have been taken advantage of in various assays using whole virions. Given the limitations of using whole virions for routine studies, a recombinant soluble molecule composed of the extracellular domain of CHE fused to the Fc region of human IgG (CHE-Fc) is engineered. Treatment of this chimeric molecule with diisopropyl fluorophosphate (DFP) selectively eliminates its esterase activity while preserving its binding property (CHE-FcD). So, this probe serves as a bifunctional tool for selectively removing (with CHE-Fc) or specifically detecting (with CHE-FcD) 9- O -acetylated sialic acids. The chapter describes the preparation of CHE-Fc chimera, chimera purification, the preparation of CHE-FcD, assay of esterase activity (CHE-Fc), CHE-FcD-binding activity assay, Use of CHE-Fc/FcD in flow cytometric analyses, and immunohistology.


Journal of Biological Chemistry | 2017

Studies on the detection, expression, glycosylation, dimerization and ligand binding properties of mouse Siglec-E

Shoib S. Siddiqui; Flavio Schwarz; Stevan A. Springer; Zahra Khedri; Hai Yu; Lingquan Deng; Andrea Verhagen; Yuko Naito-Matsui; Weiping Jiang; Daniel Kim; Jie Zhou; Beibei Ding; Xi Chen; Nissi M. Varki; Ajit Varki

CD33-related Siglecs are a family of proteins widely expressed on innate immune cells. Binding of sialylated glycans or other ligands triggers signals that inhibit or activate inflammation. Immunomodulation by Siglecs has been extensively studied, but relationships between structure and functions are poorly explored. Here we present new data relating to the structure and function of Siglec-E, the major CD33-related Siglec expressed on mouse neutrophils, monocytes, macrophages, and dendritic cells. We generated nine new rat monoclonal antibodies specific to mouse Siglec-E, with no cross-reactivity to Siglec-F. Although all antibodies detected Siglec-E on transfected human HEK-293T cells, only two reacted with mouse bone marrow neutrophils by flow cytometry and on spleen sections by immunohistochemistry. Moreover, whereas all antibodies recognized Siglec-E-Fc on immunoblots, binding was dependent on intact disulfide bonds and N-glycans, and only two antibodies recognized native Siglec-E within spleen lysates. Thus, we further investigated the impact of Siglec-E homodimerization. Homology-based structural modeling predicted a cysteine residue (Cys-298) in position to form a disulfide bridge between two Siglec-E polypeptides. Mutagenesis of Cys-298 confirmed its role in dimerization. In keeping with the high level of 9-O-acetylation found in mice, sialoglycan array studies indicate that this modification has complex effects on recognition by Siglec-E, in relationship to the underlying structures. However, we found no differences in phosphorylation or SHP-1 recruitment between dimeric and monomeric Siglec-E expressed on HEK293A cells. Phylogenomic analyses predicted that only some human and mouse Siglecs form disulfide-linked dimers. Notably, Siglec-9, the functionally equivalent human paralog of Siglec-E, occurs as a monomer.


Journal of Biological Chemistry | 2017

Physiological Exploration of the Long-term Evolutionary Selection Against Expression of N-glycolylneuraminic Acid in the Brain

Yuko Naito-Matsui; Leela R. L. Davies; Hiromu Takematsu; Hsun Hua Chou; Pam Tangvoranuntakul; Aaron F. Carlin; Andrea Verhagen; Charles J. Heyser; Seung Wan Yoo; Biswa Choudhury; James C. Paton; Adrienne W. Paton; Nissi M. Varki; Ronald L. Schnaar; Ajit Varki

All vertebrate cell surfaces display a dense glycan layer often terminated with sialic acids, which have multiple functions due to their location and diverse modifications. The major sialic acids in most mammalian tissues are N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc), the latter being derived from Neu5Ac via addition of one oxygen atom at the sugar nucleotide level by CMP-Neu5Ac hydroxylase (Cmah). Contrasting with other organs that express various ratios of Neu5Ac and Neu5Gc depending on the variable expression of Cmah, Neu5Gc expression in the brain is extremely low in all vertebrates studied to date, suggesting that neural expression is detrimental to animals. However, physiological exploration of the reasons for this long term evolutionary selection has been lacking. To explore the consequences of forced expression of Neu5Gc in the brain, we have established brain-specific Cmah transgenic mice. Such Neu5Gc overexpression in the brain resulted in abnormal locomotor activity, impaired object recognition memory, and abnormal axon myelination. Brain-specific Cmah transgenic mice were also lethally sensitive to a Neu5Gc-preferring bacterial toxin, even though Neu5Gc was overexpressed only in the brain and other organs maintained endogenous Neu5Gc expression, as in wild-type mice. Therefore, the unusually strict evolutionary suppression of Neu5Gc expression in the vertebrate brain may be explained by evasion of negative effects on neural functions and by selection against pathogens.

Collaboration


Dive into the Andrea Verhagen's collaboration.

Top Co-Authors

Avatar

Ajit Varki

University of California

View shared research outputs
Top Co-Authors

Avatar

Nissi M. Varki

University of California

View shared research outputs
Top Co-Authors

Avatar

Heinz Läubli

University of California

View shared research outputs
Top Co-Authors

Avatar

Jack D. Bui

University of California

View shared research outputs
Top Co-Authors

Avatar

Xi Chen

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hai Yu

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge