Network


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

Hotspot


Dive into the research topics where Erik Bonten is active.

Publication


Featured researches published by Erik Bonten.


The EMBO Journal | 2003

Cathepsin A regulates chaperone‐mediated autophagy through cleavage of the lysosomal receptor

Ana Maria Cuervo; Linda Mann; Erik Bonten; Alessandra d'Azzo; J. Fred Dice

Protective protein/cathepsin A (PPCA) has a serine carboxypeptidase activity of unknown physiological function. We now demonstrate that this protease activity triggers the degradation of the lysosome‐associated membrane protein type 2a (lamp2a), a receptor for chaperone‐mediated autophagy (CMA). Degrada tion of lamp2a is important because its level in the lysosomal membrane is a rate‐limiting step of CMA. Cells defective in PPCA show reduced rates of lamp2a degradation, higher levels of lamp2a and higher rates of CMA. Restoration of PPCA protease activity increases rates of lamp2a degradation, reduces levels of lysosomal lamp2a and reduces rates of CMA. PPCA associates with lamp2a on the lysosomal membrane and cleaves lamp2a near the boundary between the luminal and transmembrane domains. In addition to the well‐studied role of PPCA in targeting and protecting two lysosomal glycosidases, we have defined a role for the proteolytic activity of this multifunctional protein.


Advances in Carbohydrate Chemistry and Biochemistry | 2010

Sialidases in Vertebrates : A Family Of Enzymes Tailored For Several Cell Functions*

Eugenio Monti; Erik Bonten; Alessandra d'Azzo; Roberto Bresciani; Bruno Venerando; Giuseppe Borsani; Roland Schauer; Guido Tettamanti

This review summarizes the recent research development on vertebrate sialidase biology. Sialic acid-containing compounds play important roles in many physiological processes, including cell proliferation, apoptosis and differentiation, control of cell adhesion, immune surveillance, and clearance of plasma proteins. In this context, sialidases, the glycohydrolases that remove the terminal sialic acid at the non-reducing end of various glycoconjugates, perform an equally pivotal function. Sialidases in higher organisms are differentially expressed in cells and tissues/organs, with particular subcellular distribution and substrate specificity: they are the lysosomal (NEU1), the cytosolic (NEU2), and plasma membrane- and intracellular-associated sialidases (NEU3 and NEU4). The molecular cloning of several mammalian sialidases since 1993 has boosted research in this field. Here we summarize the results obtained since 2002, when the last general review on the molecular biology of mammalian sialidases was written. In those few years many original papers dealing with different aspects of sialidase biology have been published, highlighting the increasing relevance of these enzymes in glycobiology. Attention has also been paid to the trans-sialidases, which transfer sialic acid residues from a donor sialoconjugate to an acceptor asialo substrate. These enzymes are abundantly distributed in trypanosomes and employed to express pathogenicity, also in humans. There are structural similarities and strategic differences at the level of the active site between the mammalian sialidases and trans-sialidases. A better knowledge of these properties may permit the design of better anti-pathogen drugs.


The EMBO Journal | 1998

Transport of human lysosomal neuraminidase to mature lysosomes requires protective protein/cathepsin A

Aarnoud van der Spoel; Erik Bonten; Alessandra d'Azzo

Human lysosomal N‐acetyl‐α‐neuraminidase is deficient in two lysosomal storage disorders, sialidosis, caused by structural mutations in the neuraminidase gene, and galactosialidosis, in which a primary defect of protective protein/cathepsin A (PPCA) leads to a combined deficiency of neuraminidase and β‐D‐galactosidase. These three glycoproteins can be isolated in a high molecular weight multi‐enzyme complex, and the enzymatic activity of neuraminidase is contingent on its interaction with PPCA. To explain the unusual need of neuraminidase for an auxiliary protein, we examined, in transfected COS‐1 cells, the effect of PPCA expression on post‐translational modification, turnover and intracellular localization of neuraminidase. In pulse–chase studies, we show that the enzyme is synthesized as a 46 kDa glycoprotein, which is poorly phosphorylated, does not undergo major proteolytic processing and is secreted. Importantly, its half‐life is not altered by the presence of PPCA. However, neuraminidase associates with the PPCA precursor shortly after synthesis, since the latter protein co‐precipitates with neuraminidase using anti‐neuraminidase antibodies. We further demonstrate by subcellular fractionation of transfected cells that neuraminidase segregates to mature lysosomes only when accompanied by wild‐type PPCA, but not by transport‐impaired PPCA mutants. These data suggest a novel role for PPCA in the activation of lysosomal neuraminidase, that of an intracellular transport protein.


Journal of Leukocyte Biology | 2010

LPS-induced cytokine production in human dendritic cells is regulated by sialidase activity

Nicholas M. Stamatos; Ivan Carubelli; Diantha van de Vlekkert; Erik Bonten; Nadia Papini; Chiguang Feng; Bruno Venerando; Alessandra d'Azzo; Alan S. Cross; Lai-Xi Wang; Peter J. Gomatos

Removal of sialic acid from glycoconjugates on the surface of monocytes enhances their response to bacterial LPS. We tested the hypothesis that endogenous sialidase activity creates a permissive state for LPS‐induced cytokine production in human monocyte‐derived DCs. Of the four genetically distinct sialidases (Neu1–4), Neu1, Neu3, and Neu4 are expressed in human monocytes, but only Neu1 and Neu3 are up‐regulated as cells differentiate into DCs. Neu1 and Neu3 are present on the surface of monocytes and DCs and are also present intracellularly. DCs contain a greater amount of sialic acid than monocytes, but the amount of sialic acid/mg total protein declines during differentiation to DCs. This relative hyposialylation of cells does not occur in mature DCs grown in the presence of zanamivir, a pharmacologic inhibitor of Neu3 but not Neu1, or DANA, an inhibitor of Neu1 and Neu3. Inhibition of sialidase activity during differentiation to DCs causes no detectable change in cell viability or expression of DC surface markers. Differentiation of monocytes into DCs in the presence of zanamivir results in reduced LPS‐ induced expression of IL‐6, IL‐12p40, and TNF‐α by mature DCs, demonstrating a role for Neu3 in cytokine production. A role for Neu3 is supported by inhibition of cytokine production by DANA in DCs from Neu1–/– and WT mice. We conclude that sialidase‐mediated change in sialic acid content of specific cell surface glycoconjugates in DCs regulates LPS‐induced cytokine production, thereby contributing to development of adaptive immune responses.


Journal of Biological Chemistry | 1995

Lysosomal Protective Protein/Cathepsin A ROLE OF THE “LINKER” DOMAIN IN CATALYTIC ACTIVATION

Erik Bonten; Niels J. Galjart; Rob Willemsen; Magda Usmany; Just M. Vlak; Alessandra d'Azzo

Lysosomal protective protein/cathepsin A is a serine carboxypeptidase that forms a complex with β-galactosidase and neuraminidase. The enzyme is synthesized as a 54-kDa precursor/zymogen and processed into a catalytically active 32- and 20-kDa two-chain form. We have expressed in baculovirus-infected insect cells the human one-chain precursor as well as the two separate subunits in order to establish the mode of catalytic activation of the zymogen and the assembly and activation of the two subunits. Infected insect cells synthesize large quantities of the exogenous proteins, which are glycosylated and secreted but not processed. Co-expression of the two subunits results in their assembly into a two-chain form of 34- and 20-kDa with negligible enzymatic activity. Limited proteolysis with trypsin of the 54-kDa precursor and the reconstituted 34- and 20-kDa form gives rise to a fully active 32- and 20-kDa product. These results enabled us to map the sites of proteolytic cleavage needed for full activation of the cathepsin A zymogen. They further indicate that the 34- and 20-kDa form is a transient processing intermediate that is converted into a mature and active enzyme by removal of a 2-kDa “linker” peptide from the COOH terminus of the 34-kDa subunit.


Journal of Biological Chemistry | 2009

Heterodimerization of the Sialidase NEU1 with the Chaperone Protective Protein/Cathepsin A Prevents Its Premature Oligomerization

Erik Bonten; Yvan Campos; Viateslav Zaitsev; Amanda Nourse; Brett Waddell; William S Lewis; Garry L. Taylor; Alessandra d'Azzo

Lysosomal neuraminidase-1 (NEU1) forms a multienzyme complex with β-galactosidase and protective protein/cathepsin A (PPCA). Because of its association with PPCA, which acts as a molecular chaperone, NEU1 is transported to the lysosomal compartment, catalytically activated, and stabilized. However, the mode(s) of association between these two proteins both en route to the lysosome and in the multienzyme complex has remained elusive. Here, we have analyzed the hydrodynamic properties of PPCA, NEU1, and a complex of the two proteins and identified multiple binding sites on both proteins. One of these sites on NEU1 that is involved in binding to PPCA can also bind to other NEU1 molecules, albeit with lower affinity. Therefore, in the absence of PPCA, as in the lysosomal storage disease galactosialidosis, NEU1 self-associates into chain-like oligomers. Binding of PPCA can reverse self-association of NEU1 by causing the disassembly of NEU1-oligomers and the formation of a PPCA-NEU1 heterodimeric complex. The identification of binding sites between the two proteins allowed us to create innovative structural models of the NEU1 oligomer and the PPCA-NEU1 heterodimeric complex. The proposed mechanism of interaction between NEU1 and its accessory protein PPCA provides a rationale for the secondary deficiency of NEU1 in galactosialidosis.


Nature Genetics | 2015

NALP3 inflammasome upregulation and CASP1 cleavage of the glucocorticoid receptor cause glucocorticoid resistance in leukemia cells

Steven W. Paugh; Erik Bonten; Daniel Savic; Laura B. Ramsey; William E. Thierfelder; Prajwal Gurung; R. K. Subbarao Malireddi; Marcelo L. Actis; Anand Mayasundari; Jaeki Min; David R. Coss; Lucas T. Laudermilk; John C. Panetta; J. Robert Mccorkle; Yiping Fan; Kristine R. Crews; Gabriele Stocco; Mark R. Wilkinson; Antonio M. Ferreira; Cheng Cheng; Wenjian Yang; Seth E. Karol; Christian A. Fernandez; Barthelemy Diouf; Colton Smith; J. Kevin Hicks; Alessandra Zanut; Audrey Giordanengo; Daniel Crona; Joy J. Bianchi

Glucocorticoids are universally used in the treatment of acute lymphoblastic leukemia (ALL), and resistance to glucocorticoids in leukemia cells confers poor prognosis. To elucidate mechanisms of glucocorticoid resistance, we determined the prednisolone sensitivity of primary leukemia cells from 444 patients newly diagnosed with ALL and found significantly higher expression of CASP1 (encoding caspase 1) and its activator NLRP3 in glucocorticoid-resistant leukemia cells, resulting from significantly lower somatic methylation of the CASP1 and NLRP3 promoters. Overexpression of CASP1 resulted in cleavage of the glucocorticoid receptor, diminished the glucocorticoid-induced transcriptional response and increased glucocorticoid resistance. Knockdown or inhibition of CASP1 significantly increased glucocorticoid receptor levels and mitigated glucocorticoid resistance in CASP1-overexpressing ALL. Our findings establish a new mechanism by which the NLRP3-CASP1 inflammasome modulates cellular levels of the glucocorticoid receptor and diminishes cell sensitivity to glucocorticoids. The broad impact on the glucocorticoid transcriptional response suggests that this mechanism could also modify glucocorticoid effects in other diseases.


The FASEB Journal | 2004

Targeting macrophages with baculovirus-produced lysosomal enzymes: implications for enzyme replacement therapy of the glycoprotein storage disorder galactosialidosis

Erik Bonten; Dongning Wang; James N. Toy; Linda Mann; Aurélie Mignardot; Gouri Yogalingam; Alessandra d'Azzo

Lysosomal storage diseases (LSDs) are monogenic disorders of metabolism caused by deficiency of hydrolytic enzymes. In several LSDs, cells of the reticuloendothelial (RE) system are the primary targets of the disease. Exogenous administration of recombinant enzymes overproduced in mammalian cells has proved effective for treating the systemic phenotype in nonneuropathic patients with LSDs. However, for the treatment of diseases with primary involvement of the RE system, the production of the therapeutic enzyme in insect cells could be an alternative and advantageous method because glycoproteins expressed in insect cells carry carbohydrates of the pauci‐mannose or core‐type. These recombinant enzymes are in principle already poised to be internalized by cells that express mannose receptors, including macrophages. Here, we demonstrate that three baculovirus‐expressed enzymes, protective protein/cathepsin A (PPCA), neuraminidase (Neu1), and β‐glucosidase, were readily taken up and restored lysosomal function in enzyme‐deficient mouse macrophages. The capacity of recombinant PPCA and Neu1 to clear the lysosomal storage in target cells was assessed in PPCA−/− mice, a model of galactosialidosis. Intravenously injected PPCA−/− mice efficiently internalized the corrective enzymes in resident macrophages of many organs. In addition, treated mice showed overall clearance of lysosomal storage in the most affected systemic organs, kidney, liver, and spleen. Our results suggest that ERT with baculovirus‐expressed enzymes might be an effective treatment for nonneuropathic patients with galactosialidosis and possibly for others with LSDs that primarily involve the RE system.


Cellular and Molecular Life Sciences | 2014

Lysosomal multienzyme complex: pros and cons of working together.

Erik Bonten; Ida Annunziata; Alessandra d’Azzo

The ubiquitous distribution of lysosomes and their heterogeneous protein composition reflects the versatility of these organelles in maintaining cell homeostasis and their importance in tissue differentiation and remodeling. In lysosomes, the degradation of complex, macromolecular substrates requires the synergistic action of multiple hydrolases that usually work in a stepwise fashion. This catalytic machinery explains the existence of lysosomal enzyme complexes that can be dynamically assembled and disassembled to efficiently and quickly adapt to the pool of substrates to be processed or degraded, adding extra tiers to the regulation of the individual protein components. An example of such a complex is the one composed of three hydrolases that are ubiquitously but differentially expressed: the serine carboxypeptidase, protective protein/cathepsin A (PPCA), the sialidase, neuraminidase-1 (NEU1), and the glycosidase β-galactosidase (β-GAL). Next to this ‘core’ complex, the existence of sub-complexes, which may contain additional components, and function at the cell surface or extracellularly, suggests as yet unexplored functions of these enzymes. Here we review how studies of basic biological processes in the mouse models of three lysosomal storage disorders, galactosialidosis, sialidosis, and GM1-gangliosidosis, revealed new and unexpected roles for the three respective affected enzymes, Ppca, Neu1, and β-Gal, that go beyond their canonical degradative activities. These findings have broadened our perspective on their functions and may pave the way for the development of new therapies for these lysosomal storage disorders.


Biochemical Society Transactions | 2010

Molecular mechanisms of pathogenesis in a glycosphingolipid and a glycoprotein storage disease.

Alessandra d'Azzo; Erik Bonten

The lysosomal system comprises a specialized network of organelles crucial for the sorting, digestion, recycling and secretion of cellular components. With their content of hydrolytic enzymes, lysosomes regulate the degradation of a multitude of substrates that reach these organelles via the biosynthetic or the endocytic route. Gene defects that affect one or more of these hydrolases lead to LSDs (lysosomal storage diseases). This underscores the apparent lack of redundancy of these enzymes and the importance of the lysosomal system in cell and tissue homoeostasis. Some of the lysosomal enzymes may form multiprotein complexes, which usually work synergistically on substrates and, in this configuration, may respond more efficiently to changes in substrate load and composition. A well-characterized lysosomal multienzyme complex is the one comprising the glycosidases β-gal (β-galactosidase) and NEU1 (neuramidase-1), and of the serine carboxypeptidase PPCA (protective protein/cathepsin A). Three neurodegenerative LSDs are caused by either single or combined deficiency of these lysosomal enzymes. Sialidosis (NEU1 deficiency) and galactosialidosis (combined NEU1 and β-gal deficiency, secondary to a primary defect of PPCA) belong to the glycoprotein storage diseases, whereas GM1-gangliosidosis (β-gal deficiency) is a glycosphingolipid storage disease. Identification of novel molecular pathways that are deregulated because of loss of enzyme activity and/or accumulation of specific metabolites in various cell types has shed light on mechanisms of disease pathogenesis and may pave the way for future development of new therapies for these LSDs.

Collaboration


Dive into the Erik Bonten's collaboration.

Top Co-Authors

Avatar

Alessandra d'Azzo

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Alessandra d’Azzo

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Linda Mann

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Gouri Yogalingam

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Huimin Hu

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Diantha van de Vlekkert

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Steven W. Paugh

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Arthur W. Nienhuis

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Dongning Wang

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

E. Zanoteli

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge