Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cancer Discov ; 13(9): 2050-2071, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37272843

ABSTRACT

Most circulating tumor cells (CTC) are detected as single cells, whereas a small proportion of CTCs in multicellular clusters with stemness properties possess 20- to 100-times higher metastatic propensity than the single cells. Here we report that CTC dynamics in both singles and clusters in response to therapies predict overall survival for breast cancer. Chemotherapy-evasive CTC clusters are relatively quiescent with a specific loss of ST6GAL1-catalyzed α2,6-sialylation in glycoproteins. Dynamic hyposialylation in CTCs or deficiency of ST6GAL1 promotes cluster formation for metastatic seeding and enables cellular quiescence to evade paclitaxel treatment in breast cancer. Glycoproteomic analysis reveals newly identified protein substrates of ST6GAL1, such as adhesion or stemness markers PODXL, ICAM1, ECE1, ALCAM1, CD97, and CD44, contributing to CTC clustering (aggregation) and metastatic seeding. As a proof of concept, neutralizing antibodies against one newly identified contributor, PODXL, inhibit CTC cluster formation and lung metastasis associated with paclitaxel treatment for triple-negative breast cancer. SIGNIFICANCE: This study discovers that dynamic loss of terminal sialylation in glycoproteins of CTC clusters contributes to the fate of cellular dormancy, advantageous evasion to chemotherapy, and enhanced metastatic seeding. It identifies PODXL as a glycoprotein substrate of ST6GAL1 and a candidate target to counter chemoevasion-associated metastasis of quiescent tumor cells. This article is featured in Selected Articles from This Issue, p. 1949.


Subject(s)
Breast Neoplasms , Neoplastic Cells, Circulating , Triple Negative Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Neoplastic Cells, Circulating/metabolism , Paclitaxel/therapeutic use , Glycoproteins , Biomarkers, Tumor , Neoplasm Metastasis
2.
Anal Chem ; 90(6): 3670-3675, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29473730

ABSTRACT

Enumeration of circulating tumor cells (CTCs) of small-cell lung cancer (SCLC) patients has been shown to predict the disease progress and long-term survival. Most CTC detection methods rely on epithelial surface markers, such as epithelial cell adhesion molecule (EpCAM). However, this marker in SCLC is reported to be often downregulated after a variety of phenotypic changes, which impairs the reliability of EpCAM-based CTC detections. In this regard, the development of an alternative CTC detection method involving different CTC surface markers is in demand. In this study, we evaluated, for the first time to our knowledge, the feasibility of detecting SCLC CTCs using a noncatalytic endosialidase (EndoN Trap, EndoNt). This noncatalytic enzyme was chosen due to its high affinity to polysialic acid (polySia), a cell-surface glycan, that is highly expressed by SCLC tissue. Furthermore, this enzyme-based system was integrated into our dendrimer-mediated CTC capture platform to further enhance the capture efficiency via multivalent binding. We found that the EndoNt-immobilized surfaces could specifically capture polySia-positive SCLC cells and the binding between SCLC cells and EndoNt surfaces was further stabilized by dendrimer-mediated multivalent binding. When compared to the EpCAM-based capture, EndoNt significantly improved the capture efficiency of polySia-positive SCLC cells under flow due to its higher binding affinity (lower dissociation rate constants). These findings suggest that this enzyme-based CTC capture strategy has the potential to be used as a superior alternative to the commonly used EpCAM-based methods, particularly for those types of cancer that overexpress polySia.


Subject(s)
Cell Count/methods , Cell Separation/methods , Glycoproteins/metabolism , Lung Neoplasms/metabolism , Neoplastic Cells, Circulating/metabolism , Neuraminidase/metabolism , Small Cell Lung Carcinoma/metabolism , Cell Line , Cell Line, Tumor , Humans , Lung Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Protein Binding , Small Cell Lung Carcinoma/pathology
3.
J Biol Chem ; 293(2): 701-716, 2018 01 12.
Article in English | MEDLINE | ID: mdl-29183999

ABSTRACT

Polysialic acid (polySia) is a large glycan polymer that is added to some glycoproteins by two polysialyltransferases (polySTs), ST8Sia-II and ST8Sia-IV. As polySia modulates cell adhesion and signaling, immune cell function, and tumor metastasis, it is of interest to determine how the polySTs recognize their select substrates. We have recently identified residues within the ST8Sia-IV polybasic region (PBR) that are required for neural cell adhesion molecule (NCAM) recognition and subsequent polysialylation. Here, we compared the PBR sequence requirements for NCAM, neuropilin-2 (NRP-2), and synaptic cell adhesion molecule 1 (SynCAM 1) for polysialylation by their respective polySTs. We found that the polySTs use unique but overlapping sets of PBR residues for substrate recognition, that the NCAM-recognizing PBR sites in ST8Sia-II and ST8Sia-IV include homologous residues, but that the ST8Sia-II site is larger, and that fewer PBR residues are involved in NRP-2 and SynCAM 1 recognition than in NCAM recognition. Noting that the two sites for ST8Sia-IV autopolysialylation flank the PBR, we evaluated the role of PBR residues in autopolysialylation and found that the requirements for polyST autopolysialylation and substrate polysialylation overlap. These data together with the evaluation of the polyST autopolysialylation mechanism enabled us to further identify PBR residues potentially playing dual roles in substrate recognition and in polySia chain polymerization. Finally, we found that ST8Sia-IV autopolysialylation is required for NRP-2 polysialylation and that ST8Sia-II autopolysialylation promotes the polymerization of longer polySia chains on SynCAM 1, suggesting a critical role for polyST autopolysialylation in substrate selection and polySia chain elongation.


Subject(s)
Glycoproteins/metabolism , Animals , Cell Adhesion/physiology , Chlorocebus aethiops , N-Acetylneuraminic Acid/metabolism , Neural Cell Adhesion Molecules/metabolism , Neuropilin-2/metabolism , Sialic Acids/metabolism , Sialyltransferases/metabolism
4.
Biochemistry ; 56(10): 1504-1517, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28233978

ABSTRACT

Polysialic acid (polySia) is a unique post-translational modification found on a small set of mammalian glycoproteins. Composed of long chains of α2,8-linked sialic acid, this large, negatively charged polymer attenuates protein and cell adhesion and modulates signaling mediated by its carriers and proteins that interact with these carriers. PolySia is crucial for the proper development of the nervous system and is upregulated during tissue regeneration and in highly invasive cancers. Our laboratory has previously shown that the neural cell adhesion molecule, NCAM, has an acidic surface patch in its first fibronectin type III repeat (FN1) that is critical for the polysialylation of N-glycans on the adjacent immunoglobulin domain (Ig5). We have also identified a polysialyltransferase (polyST) polybasic region (PBR) that may mediate substrate recognition. However, a direct interaction between the NCAM FN1 acidic patch and the polyST PBR has yet to be demonstrated. Here, we have probed this interaction using isothermal titration calorimetry and nuclear magnetic resonance (NMR) spectroscopy. We observe direct and specific binding between FN1 and the PBR peptide that is dependent upon acidic residues in FN1 and basic residues of the PBR. NMR titration experiments verified the role of the FN1 acidic patch in the recognition of the PBR and suggest a conformational change of the Ig5-FN1 linker region following binding of the PBR to the acidic patch. Finally, mutation of residues identified by NMR titration experiments impacts NCAM polysialylation, supporting their mechanistic role in protein-specific polysialylation.


Subject(s)
Fibronectin Type III Domain/genetics , Neural Cell Adhesion Molecules/chemistry , Protein Processing, Post-Translational , Recombinant Fusion Proteins/chemistry , Sialic Acids/chemistry , Sialyltransferases/chemistry , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Histidine/genetics , Histidine/metabolism , Humans , Models, Molecular , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Oligopeptides/genetics , Oligopeptides/metabolism , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Binding , Protein Structure, Secondary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Sequence Alignment , Sialic Acids/metabolism , Sialyltransferases/genetics , Sialyltransferases/metabolism
5.
Histochem Cell Biol ; 147(2): 149-174, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27975143

ABSTRACT

Sialylated N-glycans play essential roles in the immune system, pathogen recognition and cancer. This review approaches the sialylation of N-glycans from three perspectives. The first section focuses on the sialyltransferases that add sialic acid to N-glycans. Included in the discussion is a description of these enzymes' glycan acceptors, conserved domain organization and sequences, molecular structure and catalytic mechanism. In addition, we discuss the protein interactions underlying the polysialylation of a select group of adhesion and signaling molecules. In the second section, the biosynthesis of sialic acid, CMP-sialic acid and sialylated N-glycans is discussed, with a special emphasis on the compartmentalization of these processes in the mammalian cell. The sequences and mechanisms maintaining the sialyltransferases and other glycosylation enzymes in the Golgi are also reviewed. In the final section, we have chosen to discuss processes in which sialylated glycans, both N- and O-linked, play a role. The first part of this section focuses on sialic acid-binding proteins including viral hemagglutinins, Siglecs and selectins. In the second half of this section, we comment on the role of sialylated N-glycans in cancer, including the roles of ß1-integrin and Fas receptor N-glycan sialylation in cancer cell survival and drug resistance, and the role of these sialylated proteins and polysialic acid in cancer metastasis.


Subject(s)
Cells/metabolism , Polysaccharides/metabolism , Sialic Acids/chemistry , Animals , Humans , Neoplasms/physiopathology , Polysaccharides/chemistry , Selectins/chemistry , Sialic Acid Binding Immunoglobulin-like Lectins/chemistry , Sialic Acids/physiology
6.
J Biol Chem ; 291(18): 9444-57, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-26884342

ABSTRACT

Polysialic acid is an oncofetal glycopolymer, added to the glycans of a small group of substrates, that controls cell adhesion and signaling. One of these substrates, neuropilin-2, is a VEGF and semaphorin co-receptor that is polysialylated on its O-glycans in mature dendritic cells and macrophages by the polysialyltransferase ST8SiaIV. To understand the biochemical basis of neuropilin-2 polysialylation, we created a series of domain swap chimeras with sequences from neuropilin-1, a protein for which polysialylation had not been previously reported. To our surprise, we found that membrane-associated neuropilin-1 is polysialylated at ∼50% of the level of neuropilin-2 but not polysialylated when it lacks its cytoplasmic tail and transmembrane region and is secreted from the cell. This was not the case for neuropilin-2, which is polysialylated when either membrane-associated or soluble. Evaluation of the soluble chimeric proteins demonstrated that the meprin A5 antigen-µ tyrosine phosphatase (MAM) domain and the O-glycan-containing linker region of neuropilin-2 are necessary and sufficient for its polysialylation and serve as better recognition and acceptor sites in the polysialylation process than those regions of neuropilin-1. In addition, specific acidic residues on the surface of the MAM domain are critical for neuropilin-2 polysialylation. Based on these data and pull-down experiments, we propose a model where ST8SiaIV recognizes and docks on an acidic surface of the neuropilin-2 MAM domain to polysialylate O-glycans on the adjacent linker region. These results together with those related to neural cell adhesion molecule polysialylation establish a paradigm for the process of protein-specific polysialylation.


Subject(s)
N-Acetylneuraminic Acid/metabolism , Neuropilin-2/metabolism , Sialyltransferases/metabolism , Animals , COS Cells , Chlorocebus aethiops , Glycosylation , Humans , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , N-Acetylneuraminic Acid/genetics , Neuropilin-2/genetics , Sialyltransferases/genetics
7.
Crit Rev Biochem Mol Biol ; 49(6): 498-532, 2014.
Article in English | MEDLINE | ID: mdl-25373518

ABSTRACT

As an anti-adhesive, a reservoir for key biological molecules, and a modulator of signaling, polysialic acid (polySia) is critical for nervous system development and maintenance, promotes cancer metastasis, tissue regeneration and repair, and is implicated in psychiatric diseases. In this review, we focus on the biosynthesis and functions of mammalian polySia, and the use of polySia in therapeutic applications. PolySia modifies a small subset of mammalian glycoproteins, with the neural cell adhesion molecule, NCAM, serving as its major carrier. Studies show that mammalian polysialyltransferases employ a unique recognition mechanism to limit the addition of polySia to a select group of proteins. PolySia has long been considered an anti-adhesive molecule, and its impact on cell adhesion and signaling attributed directly to this property. However, recent studies have shown that polySia specifically binds neurotrophins, growth factors, and neurotransmitters and that this binding depends on chain length. This work highlights the importance of considering polySia quality and quantity, and not simply its presence or absence, as its various roles are explored. The capsular polySia of neuroinvasive bacteria allows these organisms to evade the host immune response. While this "stealth" characteristic has made meningitis vaccine development difficult, it has also made polySia a worthy replacement for polyetheylene glycol in the generation of therapeutic proteins with low immunogenicity and improved circulating half-lives. Bacterial polysialyltransferases are more promiscuous than the protein-specific mammalian enzymes, and new studies suggest that these enzymes have tremendous therapeutic potential, especially for strategies aimed at neural regeneration and tissue repair.


Subject(s)
Neural Cell Adhesion Molecules/metabolism , Sialic Acids/metabolism , Sialyltransferases/metabolism , Amino Acid Sequence , Animals , Biosynthetic Pathways , Humans , Models, Molecular , Molecular Sequence Data , Neural Cell Adhesion Molecules/analysis , Neural Cell Adhesion Molecules/genetics , Sialic Acids/analysis , Sialic Acids/genetics , Sialyltransferases/analysis , Sialyltransferases/genetics
8.
Nat Chem Biol ; 10(6): 410-1, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24727902

ABSTRACT

The addition of polysialic acid to proteins and cells is emerging as a promising therapeutic strategy. Polysialyltransferases synthesize polymers of widely varying lengths not optimal for therapeutic reagents, but the development of enzyme variants using neutral genetic drift offers a new way to overcome this problem.


Subject(s)
Neisseria meningitidis, Serogroup B/enzymology , Protein Engineering , Sialyltransferases/chemistry , Sialyltransferases/genetics
9.
J Biol Chem ; 288(10): 7282-93, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23341449

ABSTRACT

The neural cell adhesion molecule (NCAM) is the major substrate for the polysialyltransferases (polySTs), ST8SiaII/STX and ST8SiaIV/PST. The polysialylation of NCAM N-glycans decreases cell adhesion and alters signaling. Previous work demonstrated that the first fibronectin type III repeat (FN1) of NCAM is required for polyST recognition and the polysialylation of the N-glycans on the adjacent Ig5 domain. In this work, we highlight the importance of an FN1 acidic patch in polyST recognition and also reveal that the polySTs are required to interact with sequences in the Ig5 domain for polysialylation to occur. We find that features of the Ig5 domain of the olfactory cell adhesion molecule (OCAM) are responsible for its lack of polysialylation. Specifically, two basic OCAM Ig5 residues (Lys and Arg) found near asparagines equivalent to those carrying the polysialylated N-glycans in NCAM substantially decrease or eliminate polysialylation when used to replace the smaller and more neutral residues (Ser and Asn) in analogous positions in NCAM Ig5. This decrease in polysialylation does not reflect altered glycosylation but instead is correlated with a decrease in polyST-NCAM binding. In addition, inserting non-conserved OCAM sequences into NCAM Ig5, including an "extra" N-glycosylation site, decreases or completely blocks NCAM polysialylation. Taken together, these results indicate that the polySTs not only recognize an acidic patch in the FN1 domain of NCAM but also must contact sequences in the Ig5 domain for polysialylation of Ig5 N-glycans to occur.


Subject(s)
Neural Cell Adhesion Molecules/metabolism , Polysaccharides/metabolism , Sialic Acids/metabolism , Sialyltransferases/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , COS Cells , Chlorocebus aethiops , Fibronectins/genetics , Fibronectins/metabolism , Glycosylation , Immunoblotting , Immunoglobulins/genetics , Immunoglobulins/metabolism , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Mutation , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/genetics , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Sialyltransferases/genetics , Substrate Specificity
10.
J Biol Chem ; 287(9): 6441-53, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22184126

ABSTRACT

Polysialic acid on the neural cell adhesion molecule (NCAM) modulates cell-cell adhesion and signaling, is required for proper brain development, and plays roles in neuronal regeneration and the growth and invasiveness of tumor cells. Evidence indicates that NCAM polysialylation is highly protein-specific, requiring an initial polysialyltransferase-NCAM protein-protein interaction. Previous work suggested that a polybasic region located prior to the conserved polysialyltransferase catalytic motifs may be involved in NCAM recognition, but not overall enzyme activity (Foley, D. A., Swartzentruber, K. G., and Colley, K. J. (2009) J. Biol. Chem. 284, 15505-15516). Here, we employ a competition assay to evaluate the role of this region in substrate recognition. We find that truncated, catalytically inactive ST8SiaIV/PST proteins that include the polybasic region, but not those that lack this region, compete with endogenous ST8SiaIV/PST and reduce NCAM polysialylation in SW2 small cell lung carcinoma cells. Replacing two polybasic region residues, Arg(82) and Arg(93), eliminates the ability of a full-length, catalytically inactive enzyme (PST H331K) to compete with SW2 cell ST8SiaIV/PST and block NCAM polysialylation. Replacing these residues singly or together in ST8SiaIV/PST substantially reduces or eliminates NCAM polysialylation, respectively. In contrast, replacing Arg(82), but not Arg(93), substantially reduces the ability of ST8SiaIV/PST to polysialylate neuropilin-2 and SynCAM 1, suggesting that Arg(82) plays a general role in substrate recognition, whereas Arg(93) specifically functions in NCAM recognition. Taken together, our results indicate that the ST8SiaIV/PST polybasic region plays a critical role in substrate recognition and suggest that different combinations of basic residues may mediate the recognition of distinct substrates.


Subject(s)
Neural Cell Adhesion Molecule L1/metabolism , Sialic Acids/metabolism , Sialyltransferases/genetics , Sialyltransferases/metabolism , Animals , Arginine/metabolism , CHO Cells , COS Cells , Catalytic Domain/genetics , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/metabolism , Chlorocebus aethiops , Conserved Sequence , Cricetinae , Humans , Immunoglobulins/metabolism , Lung Neoplasms , Mutagenesis/physiology , N-Acetylneuraminic Acid/metabolism , Neuropilin-2/metabolism , Protein Structure, Tertiary/physiology , Sialyltransferases/chemistry , Small Cell Lung Carcinoma , Substrate Specificity/genetics
11.
J Biol Chem ; 286(6): 4525-34, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21131353

ABSTRACT

Polysialic acid is an anti-adhesive glycan that modifies a select group of mammalian proteins. The primary substrate of the polysialyltransferases (polySTs) is the neural cell adhesion molecule (NCAM). Polysialic acid negatively regulates cell adhesion, is required for proper brain development, and is expressed in specific areas of the adult brain where it promotes on-going cell migration and synaptic plasticity. The first fibronectin type III repeat (FN1) of NCAM is required for polysialylation of the N-glycans on the adjacent immunoglobulin-like domain (Ig5), and acidic residues on the surface of FN1 play a role in polyST recognition. Recent work demonstrated that the FN1 domain from the unpolysialylated olfactory cell adhesion molecule (OCAM) was able to partially replace NCAM FN1 (Foley, D. A., Swartzentruber, K. G., Thompson, M. G., Mendiratta, S. S., and Colley, K. J. (2010) J. Biol. Chem. 285, 35056-35067). Here we demonstrate that individually replacing three identical regions shared by NCAM and OCAM FN1, (500)PSSP(503) (PSSP), (526)GGVPI(530) (GGVPI), and (580)NGKG(583) (NGKG), dramatically reduces NCAM polysialylation. In addition, we show that the polyST, ST8SiaIV/PST, specifically binds NCAM and that this binding requires the FN1 domain. Replacing the FN1 PSSP sequences and the acidic patch residues decreases NCAM-polyST binding, whereas replacing the GGVPI and NGKG sequences has no effect. The location of GGVPI and NGKG in loops that flank the Ig5-FN1 linker and the proximity of PSSP to this linker suggest that GGVPI and NGKG sequences may be critical for stabilizing the Ig5-FN1 linker, whereas PSSP may play a dual role maintaining the Ig5-FN1 interface and a polyST recognition site.


Subject(s)
N-Acetylneuraminic Acid/metabolism , Neural Cell Adhesion Molecules/metabolism , Protein Processing, Post-Translational/physiology , Animals , COS Cells , Chlorocebus aethiops , Dogs , Humans , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/genetics , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Repetitive Sequences, Amino Acid
12.
J Biol Chem ; 285(45): 35056-67, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20805222

ABSTRACT

Polysialic acid is a developmentally regulated, anti-adhesive polymer that is added to N-glycans on the fifth immunoglobulin domain (Ig5) of the neural cell adhesion molecule (NCAM). We found that the first fibronectin type III repeat (FN1) of NCAM is required for the polysialylation of N-glycans on the adjacent Ig5 domain, and we proposed that the polysialyltransferases recognize specific sequences in FN1 to position themselves for Ig5 N-glycan polysialylation. Other studies identified a novel FN1 acidic surface patch and α-helix that play roles in NCAM polysialylation. Here, we characterize the contribution of two additional FN1 sequences, Pro(510)-Tyr(511)-Ser(512) (PYS) and Gln(516)-Val(517)-Gln(518) (QVQ). Replacing PYS or the acidic patch dramatically decreases the O-glycan polysialylation of a truncated NCAM protein, and replacing the α-helix or QVQ shifts polysialic acid to FN1 O-glycans in full-length NCAM. We also found that the FN1 domain of the olfactory cell adhesion molecule, a homologous but unpolysialylated protein, could partially replace NCAM FN1. Inserting Pro(510)-Tyr(511) eliminated N-glycan polysialylation and enhanced O-glycosylation of an NCAM- olfactory cell adhesion molecule chimera, and inserting other FN1 sequences unique to NCAM, predominantly the acidic patch, created a new polysialyltransferase recognition site. Taken together, our results highlight the role of the FN1 α-helix and QVQ sequences in N-glycan polysialylation and demonstrate that the acidic patch primarily functions in O-glycan polysialylation.


Subject(s)
Fibronectins , N-Acetylneuraminic Acid/metabolism , Neural Cell Adhesion Molecules/metabolism , Polysaccharides/metabolism , Animals , COS Cells , Chlorocebus aethiops , Humans , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/genetics , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/genetics , Polysaccharides/chemistry , Polysaccharides/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sialyltransferases/genetics , Sialyltransferases/metabolism
13.
J Biol Chem ; 285(35): 27360-27371, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20573953

ABSTRACT

The addition of alpha2,8-polysialic acid to the N-glycans of the neural cell adhesion molecule, NCAM, is critical for brain development and plays roles in synaptic plasticity, learning and memory, neuronal regeneration, and the growth and invasiveness of cancer cells. Our previous work indicates that the polysialylation of two N-glycans located on the fifth immunoglobulin domain (Ig5) of NCAM requires the presence of specific sequences in the adjacent fibronectin type III repeat (FN1). To understand the relationship of these two domains, we have solved the crystal structure of the NCAM Ig5-FN1 tandem. Unexpectedly, the structure reveals that the sites of Ig5 polysialylation are on the opposite face from the FN1 residues previously found to be critical for N-glycan polysialylation, suggesting that the Ig5-FN1 domain relationship may be flexible and/or that there is flexibility in the placement of Ig5 glycosylation sites for polysialylation. To test the latter possibility, new Ig5 glycosylation sites were engineered and their polysialylation tested. We observed some flexibility in glycosylation site location for polysialylation and demonstrate that the lack of polysialylation of a glycan attached to Asn-423 may be in part related to a lack of terminal processing. The data also suggest that, although the polysialyltransferases do not require the Ig5 domain for NCAM recognition, their ability to engage with this domain is necessary for polysialylation to occur on Ig5 N-glycans.


Subject(s)
Cell Adhesion Molecules, Neuronal/chemistry , Sialic Acids/chemistry , Amino Acid Motifs , Animals , COS Cells , Cell Adhesion Molecules, Neuronal/biosynthesis , Cell Adhesion Molecules, Neuronal/genetics , Chlorocebus aethiops , Humans , Protein Structure, Tertiary , Sialic Acids/genetics , Sialic Acids/metabolism , Structure-Activity Relationship
15.
J Biol Chem ; 284(23): 15505-16, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19336400

ABSTRACT

The polysialyltransferases ST8Sia II and ST8Sia IV polysialylate the glycans of a small subset of mammalian proteins. Their most abundant substrate is the neural cell adhesion molecule (NCAM). An acidic surface patch and a novel alpha-helix in the first fibronectin type III repeat of NCAM are required for the polysialylation of N-glycans on the adjacent immunoglobulin domain. Inspection of ST8Sia IV sequences revealed two conserved polybasic regions that might interact with the NCAM acidic patch or the growing polysialic acid chain. One is the previously identified polysialyltransferase domain (Nakata, D., Zhang, L., and Troy, F. A. (2006) Glycoconj. J. 23, 423-436). The second is a 35-amino acid polybasic region that contains seven basic residues and is equidistant from the large sialyl motif in both polysialyltransferases. We replaced these basic residues to evaluate their role in enzyme autopolysialylation and NCAM-specific polysialylation. We found that replacement of Arg(276)/Arg(277) or Arg(265) in the polysialyltransferase domain of ST8Sia IV decreased both NCAM polysialylation and autopolysialylation in parallel, suggesting that these residues are important for catalytic activity. In contrast, replacing Arg(82)/Arg(93) in ST8Sia IV with alanine substantially decreased NCAM-specific polysialylation while only partially impacting autopolysialylation, suggesting that these residues may be particularly important for NCAM polysialylation. Two conserved negatively charged residues, Glu(92) and Asp(94), surround Arg(93). Replacement of these residues with alanine largely inactivated ST8Sia IV, whereas reversing these residues enhanced enzyme autopolysialylation but significantly reduced NCAM polysialylation. In sum, we have identified selected amino acids in this conserved polysialyltransferase polybasic region that are critical for the protein-specific polysialylation of NCAM.


Subject(s)
Neural Cell Adhesion Molecules/metabolism , Sialyltransferases/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Arginine , COS Cells , Chlorocebus aethiops , Humans , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , N-Acetylneuraminic Acid/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sialyltransferases/chemistry , Sialyltransferases/genetics , Substrate Specificity , Transfection
16.
J Biol Chem ; 281(47): 36052-9, 2006 Nov 24.
Article in English | MEDLINE | ID: mdl-17003032

ABSTRACT

Polysialic acid is a developmentally regulated, anti-adhesive glycan that is added to the neural cell adhesion molecule, NCAM. Polysialylated NCAM is critical for brain development and plays roles in synaptic plasticity, axon guidance, and cell migration. The first fibronectin type III repeat of NCAM, FN1, is necessary for the polysialylation of N-glycans on the adjacent immunoglobulin domain. This repeat cannot be replaced by other fibronectin type III repeats. We solved the crystal structure of human NCAM FN1 and found that, in addition to a unique acidic surface patch, it possesses a novel alpha-helix that links strands 4 and 5 of its beta-sandwich structure. Replacement of the alpha-helix did not eliminate polysialyltransferase recognition, but shifted the addition of polysialic acid from the N-glycans modifying the adjacent immunoglobulin domain to O-glycans modifying FN1. Other experiments demonstrated that replacement of residues in the acidic surface patch alter the polysialylation of both N- and O-glycans in the same way, while the alpha-helix is only required for the polysialylation of N-glycans. Our data are consistent with a model in which the FN1 alpha-helix is involved in an Ig5-FN1 interaction that is critical for the correct positioning of Ig5 N-glycans for polysialylation.


Subject(s)
Fibronectins/chemistry , Neural Cell Adhesion Molecules/chemistry , Animals , COS Cells , Chlorocebus aethiops , Crystallography, X-Ray , DNA, Complementary/metabolism , Humans , Models, Molecular , Polysaccharides/chemistry , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Sialic Acids/chemistry , Sialic Acids/metabolism , Sialyltransferases/chemistry
17.
J Biol Chem ; 281(41): 31106-18, 2006 Oct 13.
Article in English | MEDLINE | ID: mdl-16923816

ABSTRACT

The addition of sialic acid to glycoproteins and glycolipids requires Golgi sialyltransferases to have access to their glycoconjugate substrates and nucleotide sugar donor, CMP-sialic acid. CMP-sialic acid is transported into the lumen of the Golgi complex through the CMP-sialic acid transporter, an antiporter that also functions to transport CMP into the cytosol. We localized the transporter using immunofluorescence and deconvolution microscopy to test the prediction that it is broadly distributed across the Golgi stack to serve the many sialyltransferases involved in glycoconjugate sialylation. The transporter co-localized with ST6GalI in the medial and trans Golgi, showed partial overlap with a medial Golgi marker and little overlap with early Golgi or trans Golgi network markers. Endoplasmic reticulum-retained forms of sialyltransferases did not redistribute the transporter from the Golgi to the endoplasmic reticulum, suggesting that transporter-sialyltransferase complexes are not involved in transporter localization. Next we evaluated the role of the transporter's N- and C-terminal cytoplasmic tails in its trafficking and localization. The N-tail was not required for either endoplasmic reticulum export or Golgi localization. The C-tail was required for endoplasmic reticulum export and contained di-Ile and terminal Val motifs at its very C terminus that function as independent endoplasmic reticulum export signals. Deletion of the last four amino acids of the C-tail (IIGV) eliminated these export signals and prevented endoplasmic reticulum export of the transporter. This form of the transporter supplied limited amounts of CMP-sialic acid to Golgi sialyltransferases but was unable to completely rescue the transporter defect of Lec2 Chinese hamster ovary cells.


Subject(s)
Antiporters/physiology , Cytidine Monophosphate N-Acetylneuraminic Acid/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Amino Acid Sequence , Animals , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , HeLa Cells , Humans , Mice , Molecular Sequence Data , Sialyltransferases/metabolism , beta-D-Galactoside alpha 2-6-Sialyltransferase
18.
J Biol Chem ; 280(37): 32340-8, 2005 Sep 16.
Article in English | MEDLINE | ID: mdl-16027151

ABSTRACT

Polysialic acid is an anti-adhesive protein modification that promotes cell migration and the plasticity of cell interactions. Because so few proteins carry polysialic acid, we hypothesized that polysialylation is a protein-specific event and that a specific polysialyltransferase-substrate interaction is the basis of this specificity. The major substrate for the polysialyltransferases is the neural cell adhesion molecule, NCAM. Previous work demonstrates that the first fibronectin type III repeat of NCAM (FN1) was necessary for the polysialylation of the N-glycans on the adjacent immunoglobulin domain (Ig5) (Close, B. E., Mendiratta, S. S., Geiger, K. M., Broom, L. J., Ho, L. L., and Colley, K. J. (2003) J. Biol. Chem. 278, 30796-30805). This suggested that FN1 may be a recognition site for the polysialyltransferases. In this study, we showed that the second fibronectin type III repeat (FN2) of NCAM cannot replace FN1. Arg substitution of three unique acidic amino acids on the surface of FN1 eliminated polysialylation not only of a minimal Ig5-FN1 substrate but also of full-length NCAM. Ala substitution of these residues eliminated Ig5-FN1 polysialylation but not that of full-length NCAM, suggesting that the two proteins are interacting differently with the enzymes and that multiple residues are involved in the enzyme-NCAM interaction. By using another truncated protein, Ig5-FN1-FN2, we confirmed the importance of enzyme-substrate positioning for optimal recognition and polysialylation. In sum, we have found that acidic residues on the surface of FN1 are part of a larger protein interaction region that is critical for NCAM recognition and polysialylation by the polysialyltransferases.


Subject(s)
Fibronectins/chemistry , N-Acetylneuraminic Acid/metabolism , Neurons/metabolism , Amino Acid Sequence , Amino Acids/chemistry , Animals , Binding Sites , COS Cells , Cell Adhesion , DNA, Complementary/metabolism , Epitopes/chemistry , Fluorescent Antibody Technique, Indirect , Gene Deletion , Humans , Magnetic Resonance Spectroscopy , Microscopy, Fluorescence , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Protein Structure, Tertiary , Sialic Acids/chemistry , Sialyltransferases/chemistry , Substrate Specificity , Transfection
19.
J Biol Chem ; 280(7): 5423-9, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15582997

ABSTRACT

A single amino acid difference in the catalytic domain of two isoforms of the alpha2,6-sialyltransferase (ST6Gal I) leads to differences in their trafficking, processing, and oligomerization. The STtyr isoform is transiently localized in the Golgi and is ultimately cleaved and secreted, whereas the STcys isoform is stably localized in the Golgi and is not cleaved and secreted. The stable localization of STcys is correlated with its enhanced ability to oligomerize. To test the hypothesis that multiple signals can mediate Golgi localization and further evaluate the role of oligomerization in the localization process, we evaluated the effects of individually and simultaneously altering the cytosolic tail and transmembrane region of the STcys isoform. We found that the localization, processing, and oligomerization of STcys were not substantially changed when either the core amino acids of the cytosolic tail were deleted or the sequence and length of the transmembrane region were altered. In contrast, when these changes were made simultaneously, the STcys isoform was converted into a form that was processed, secreted, and weakly oligomerized like STtyr. We propose that STcys oligomerization is a secondary event resulting from its concentration in the Golgi via mechanisms independently mediated by its cytosolic tail and transmembrane region.


Subject(s)
Golgi Apparatus/metabolism , Sialyltransferases/chemistry , Sialyltransferases/metabolism , Animals , COS Cells , Catalytic Domain , HeLa Cells , Humans , Hydrogen-Ion Concentration , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Quaternary , Sequence Deletion/genetics , Sialyltransferases/genetics , Signal Transduction , Solubility , beta-D-Galactoside alpha 2-6-Sialyltransferase
20.
J Biol Chem ; 278(33): 30796-805, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12791681

ABSTRACT

A limited number of mammalian proteins are modified by polysialic acid, with the neural cell adhesion molecule (NCAM) being the most abundant of these. We hypothesize that polysialylation is a protein-specific glycosylation event and that an initial protein-protein interaction between polysialyltransferases and glycoprotein substrates mediates this specificity. To evaluate the regions of NCAM required for recognition and polysialylation by PST/ST8Sia IV and STX/ST8Sia II, a series of domain deletion proteins were generated, co-expressed with each enzyme, and their polysialylation analyzed. A protein consisting of the fifth immunoglobulin-like domain (Ig5), which contains the reported sites of polysialylation, and the first fibronectin type III repeat (FN1) was polysialylated by both enzymes, whereas a protein consisting of Ig5 alone was not polysialylated by either enzyme. This demonstrates that the Ig5 domain of NCAM and FN1 are sufficient for polysialylation, and suggests that the FN1 may constitute an enzyme recognition and docking site. Two other NCAM mutants, NCAM-6 (Ig1-5) and NCAM-7 (FN1-FN2), were weakly polysialylated by PST/ST8Sia IV, suggesting that a weaker enzyme recognition site may exist within the Ig domains, and that glycans in the FN region are polysialylated. Further analysis indicated that O-linked oligosaccharides in NCAM-7, and O-linked and N-linked glycans in full-length NCAM, are polysialylated when these proteins are co-expressed with the polysialyltransferases in COS-1 cells. Our data support a model in which the polysialyltransferases bind to the FN1 of NCAM to polymerize polysialic acid chains on appropriately presented glycans in adjacent regions.


Subject(s)
Neural Cell Adhesion Molecule L1/chemistry , Neural Cell Adhesion Molecule L1/metabolism , Sialic Acids/chemistry , Sialic Acids/metabolism , Amino Acid Sequence , Cell Membrane/metabolism , Gene Deletion , Humans , Molecular Sequence Data , Mutagenesis , Neural Cell Adhesion Molecule L1/genetics , Protein Structure, Tertiary , Sialic Acids/genetics , Sialyltransferases/metabolism , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...