Network


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

Hotspot


Dive into the research topics where Flavio Schwarz is active.

Publication


Featured researches published by Flavio Schwarz.


Current Opinion in Structural Biology | 2011

Mechanisms and principles of N-linked protein glycosylation

Flavio Schwarz; Markus Aebi

N-linked glycosylation, a protein modification system present in all domains of life, is characterized by a high structural diversity of N-linked glycans found among different species and by a large number of proteins that are glycosylated. Based on structural, functional, and phylogenetic approaches, this review discusses the highly conserved processes that are at the basis of this unique general protein modification system.


Nature Chemical Biology | 2010

A combined method for producing homogeneous glycoproteins with eukaryotic N-glycosylation

Flavio Schwarz; Wei Huang; Cishan Li; Benjamin L. Schulz; Christian Lizak; Alessandro Palumbo; Shin Numao; Dario Neri; Markus Aebi; Lai-Xi Wang

We describe a novel method for producing homogeneous eukaryotic N-glycoproteins. The method involves the engineering and functional transfer of the C. jejuni glycosylation machinery in E. coli to express glycosylated proteins with the key GlcNAc-Asn linkage. The bacterial glycans were then trimmed and remodeled in vitro by enzymatic transglycosylation to fulfill a eukaryotic N-glycosylation. It provides a potentially general platform for producing eukaryotic N-glycoproteins.


PLOS Pathogens | 2011

Galactosaminogalactan, a New Immunosuppressive Polysaccharide of Aspergillus fumigatus

Thierry Fontaine; Aurélie Delangle; Catherine Simenel; Bernadette Coddeville; Sandra J. van Vliet; Yvette van Kooyk; Silvia Bozza; Silvia Moretti; Flavio Schwarz; Coline Trichot; Markus Aebi; Muriel Delepierre; Carole Elbim; Luigina Romani; Jean-Paul Latgé

A new polysaccharide secreted by the human opportunistic fungal pathogen Aspergillus fumigatus has been characterized. Carbohydrate analysis using specific chemical degradations, mass spectrometry, 1H and 13C nuclear magnetic resonance showed that this polysaccharide is a linear heterogeneous galactosaminogalactan composed of α1-4 linked galactose and α1-4 linked N-acetylgalactosamine residues where both monosacharides are randomly distributed and where the percentage of galactose per chain varied from 15 to 60%. This polysaccharide is antigenic and is recognized by a majority of the human population irrespectively of the occurrence of an Aspergillus infection. GalNAc oligosaccharides are an essential epitope of the galactosaminogalactan that explains the universal antibody reaction due to cross reactivity with other antigenic molecules containing GalNAc stretches such as the N-glycans of Campylobacter jejuni. The galactosaminogalactan has no protective effect during Aspergillus infections. Most importantly, the polysaccharide promotes fungal development in immunocompetent mice due to its immunosuppressive activity associated with disminished neutrophil infiltrates.


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.


Glycobiology | 2011

Relaxed acceptor site specificity of bacterial oligosaccharyltransferase in vivo

Flavio Schwarz; Christian Lizak; Yao-Yun Fan; Susanna Fleurkens; Markus Aebi

A number of proteobacteria carry the genetic information to perform N-linked glycosylation, but only the protein glycosylation (pgl) pathway of Campylobacter jejuni has been studied to date. Here, we report that the pgl gene cluster of Campylobacter lari encodes for a functional glycosylation machinery that can be reconstituted in Escherichia coli. We determined that the N-glycan produced in this system consisted of a linear hexasaccharide. We found that the oligosaccharyltransferase (OST) of C. lari conserved a predominant specificity for the primary sequence D/E-X(-1)-N-X(+1)-S/T (where X(-1) and X(+1) can be any amino acid but proline). At the same time, we observed that this enzyme exhibited a relaxed specificity toward the acceptor site and modified asparagine residues of a protein at sequences DANSG and NNNST. Moreover, C. lari pgl glycosylated a native E. coli protein. Bacterial N-glycosylation appears as a useful tool to establish a molecular description of how single-subunit OSTs perform selection of glycosyl acceptor sites.


Journal of Biological Chemistry | 2011

Cytoplasmic N-Glycosyltransferase of Actinobacillus pleuropneumoniae Is an Inverting Enzyme and Recognizes the NX(S/T) Consensus Sequence

Flavio Schwarz; Yao-Yun Fan; Mario Schubert; Markus Aebi

N-Linked glycosylation is a frequent protein modification that occurs in all three domains of life. This process involves the transfer of a preassembled oligosaccharide from a lipid donor to asparagine side chains of polypeptides and is catalyzed by the membrane-bound oligosaccharyltransferase (OST). We characterized an alternative bacterial pathway wherein a cytoplasmic N-glycosyltransferase uses nucleotide-activated monosaccharides as donors to modify asparagine residues of peptides and proteins. N-Glycosyltransferase is an inverting glycosyltransferase and recognizes the NX(S/T) consensus sequence. It therefore exhibits similar acceptor site specificity as eukaryotic OST, despite the unrelated predicted structural architecture and the apparently different catalytic mechanism. The identification of an enzyme that integrates some of the features of OST in a cytoplasmic pathway defines a novel class of N-linked protein glycosylation found in pathogenic bacteria.


Journal of Biological Chemistry | 2014

Molecular Analysis of an Alternative N-Glycosylation Machinery by Functional Transfer from Actinobacillus pleuropneumoniae to Escherichia coli

Andreas Naegeli; Christine Neupert; Yao-Yun Fan; Chia-Wei Lin; Kristina Poljak; Anna Maria Papini; Flavio Schwarz; Markus Aebi

Background: Actinobacillus pleuropneumoniae N-glycosyltransferase is a cytoplasmic glycosyltransferase catalyzing N-glycosylation of polypeptides. Results: In depth analysis of a reconstituted A. pleuropneumoniae glycosylation system in Escherichia coli showed a surprisingly relaxed peptide substrate specificity of N-glycosyltransferase. Conclusion: N-Glycosyltransferase constitutes a general glycosylation system with a preference for autotransporters. Significance: Our study could provide the basis for a novel route for the engineering of N-glycoproteins in bacteria. N-Linked protein glycosylation is a frequent post-translational modification that can be found in all three domains of life. In a canonical, highly conserved pathway, an oligosaccharide is transferred by a membrane-bound oligosaccharyltransferase from a lipid donor to asparagines in the sequon NX(S/T) of secreted polypeptides. The δ-proteobacterium Actinobacillus pleuropneumoniae encodes an unusual pathway for N-linked protein glycosylation. This pathway takes place in the cytoplasm and is mediated by a soluble N-glycosyltransferase (NGT) that uses nucleotide-activated monosaccharides to glycosylate asparagine residues. To characterize the process of cytoplasmic N-glycosylation in more detail, we studied the glycosylation in A. pleuropneumoniae and functionally transferred the glycosylation system to Escherichia coli. N-Linked glucose specific human sera were used for the analysis of the glycosylation process. We identified autotransporter adhesins as the preferred protein substrate of NGT in vivo, and in depth analysis of the modified sites in E. coli revealed a surprisingly relaxed peptide substrate specificity. Although NX(S/T) is the preferred acceptor sequon, we detected glycosylation of alternative sequons, including modification of glutamine and serine residues. We also demonstrate the use of NGT to glycosylate heterologous proteins. Therefore, our study could provide the basis for a novel route for the engineering of N-glycoproteins in bacteria.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Human-specific derived alleles of CD33 and other genes protect against postreproductive cognitive decline

Flavio Schwarz; Stevan A. Springer; Tasha K. Altheide; Nissi M. Varki; Pascal Gagneux; Ajit Varki

Significance Most vertebrates die soon after they stop reproducing, but humans are an exception. Postreproductive humans care for offspring, assist in foraging, and communicate ecological and cultural knowledge, increasing the survival of younger individuals. Loss of cognitive capacity disrupts these benefits and burdens the group with the care of older members. We studied how the immunoregulatory receptor CD33 contributes to Alzheimer’s disease, a human-specific postreproductive condition. Surprisingly, a protective CD33 allele is derived and unique to humans, despite weak direct selection on older individuals. We identified several genes with derived alleles that protect against neurodegenerative disease and cerebrovascular insufficiency in old age. Selection by inclusive fitness may be strong enough to favor alleles that protect against cognitive decline in postreproductive humans. The individuals of most vertebrate species die when they can no longer reproduce. Humans are a rare exception, having evolved a prolonged postreproductive lifespan. Elders contribute to cooperative offspring care, assist in foraging, and communicate important ecological and cultural knowledge, increasing the survival of younger individuals. Age-related deterioration of cognitive capacity in humans compromises these benefits and also burdens the group with socially costly members. We investigated the contribution of the immunoregulatory receptor CD33 to a uniquely human postreproductive disease, Alzheimer’s dementia. Surprisingly, even though selection at advanced age is expected to be weak, a CD33 allele protective against Alzheimer’s disease is derived and unique to humans and favors a functional molecular state of CD33 resembling that of the chimpanzee. Thus, derived alleles may be compensatory and restore interactions altered as a consequence of human-specific brain evolution. We found several other examples of derived alleles at other human loci that protect against age-related cognitive deterioration arising from neurodegenerative disease or cerebrovascular insufficiency. Selection by inclusive fitness may be strong enough to favor alleles protecting specifically against cognitive decline in postreproductive humans. Such selection would operate by maximizing the contributions of postreproductive individuals to the fitness of younger kin.


eLife | 2015

Siglec receptors impact mammalian lifespan by modulating oxidative stress.

Flavio Schwarz; Oliver M. T. Pearce; Xiaoxia Wang; Annie N. Samraj; Heinz Läubli; Javier O. Garcia; Hongqiao Lin; Xiaoming Fu; Andrea Garcia-Bingman; Patrick Secrest; Casey E. Romanoski; Charles J. Heyser; Christopher K. Glass; Stanley L. Hazen; Nissi M. Varki; Ajit Varki; Pascal Gagneux

Aging is a multifactorial process that includes the lifelong accumulation of molecular damage, leading to age-related frailty, disability and disease, and eventually death. In this study, we report evidence of a significant correlation between the number of genes encoding the immunomodulatory CD33-related sialic acid-binding immunoglobulin-like receptors (CD33rSiglecs) and maximum lifespan in mammals. In keeping with this, we show that mice lacking Siglec-E, the main member of the CD33rSiglec family, exhibit reduced survival. Removal of Siglec-E causes the development of exaggerated signs of aging at the molecular, structural, and cognitive level. We found that accelerated aging was related both to an unbalanced ROS metabolism, and to a secondary impairment in detoxification of reactive molecules, ultimately leading to increased damage to cellular DNA, proteins, and lipids. Taken together, our data suggest that CD33rSiglecs co-evolved in mammals to achieve a better management of oxidative stress during inflammation, which in turn reduces molecular damage and extends lifespan. DOI: http://dx.doi.org/10.7554/eLife.06184.001


Advances in Experimental Medicine and Biology | 2015

Human-Specific Evolutionary Changes in the Biology of Siglecs

Flavio Schwarz; Jerry J. Fong; Ajit Varki

Siglecs are a family of sialic acid-recognizing immunoglobulin-like lectins that exhibit multiple human-specific and human-universal differences, including changes in binding specificity (Siglec-5, -7, -9, -11, -12 and 14); changes in expression pattern (Siglec-1, -5, -6, and -11); gene conversion (SIGLEC11); gene deletion (SIGLEC13) and pseudogenization (SIGLEC17). Human-unique pseudogenes of SIGLEC12, SIGLEC14 and SIGLEC16 are also polymorphic within human populations, suggesting ongoing selection on this family of genes. The apparently higher concentration of SIGLEC changes in the human lineage may have been selected by interactions with pathogens binding Siglecs, and/or as compensatory responses to the loss of the sialic acid N-glycolylneuraminic acid (Neu5Gc) in humans. Human-specific Siglec changes of particular interest include expression of Siglec-11 in brain microglia, expression of Siglec-6 on placental trophoblast, suppression of Siglec-5 expression on adaptive immune cells, new expression of Siglec-5 on amniotic epithelium, and elimination of Siglec-13 and -17 from innate immune cells. The Siglec-13 and -17 inactivation events fixed in the ancestral population shortly before the common ancestor of modern humans 100–200 thousand years ago, and resurrected Siglec-13 and -17 gene products bind potentially lethal pathogens of infants. While such pathogens may have contributed to population bottlenecks in human evolution, the resulting changes in sialic acid biology may also have altered multiple systems where sialic acid and Siglecs have endogenous roles. Thus, genes associated with sialic acid biology appear to be a “hot spot” of genetic and physiological change during human evolution, with implications for human origins, and for uniquely human features in health and disease.

Collaboration


Dive into the Flavio Schwarz'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
Top Co-Authors

Avatar

Lingquan Deng

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heinz Läubli

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pascal Gagneux

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge