51
|
Foote CA, Castorena-Gonzalez JA, Ramirez-Perez FI, Jia G, Hill MA, Reyes-Aldasoro CC, Sowers JR, Martinez-Lemus LA. Arterial Stiffening in Western Diet-Fed Mice Is Associated with Increased Vascular Elastin, Transforming Growth Factor-β, and Plasma Neuraminidase. Front Physiol 2016; 7:285. [PMID: 27458385 PMCID: PMC4935726 DOI: 10.3389/fphys.2016.00285] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/23/2016] [Indexed: 01/06/2023] Open
Abstract
Consumption of excess fat and carbohydrate (Western diet, WD) is associated with alterations in the structural characteristics of blood vessels. This vascular remodeling contributes to the development of cardiovascular disease, particularly as it affects conduit and resistance arteries. Vascular remodeling is often associated with changes in the elastin-rich internal elastic lamina (IEL) and the activation of transforming growth factor (TGF)-β. In addition, obesity and type II diabetes have been associated with increased serum neuraminidase, an enzyme known to increase TGF-β cellular output. Therefore, we hypothesized that WD-feeding would induce structural modifications to the IEL of mesenteric resistance arteries in mice, and that these changes would be associated with increased levels of circulating neuraminidase and the up-regulation of elastin and TGF-β in the arterial wall. To test this hypothesis, a WD, high in fat and sugar, was used to induce obesity in mice, and the effect of this diet on the structure of mesenteric resistance arteries was investigated. 4-week old, Post-weaning mice were fed either a normal diet (ND) or WD for 16 weeks. Mechanically, arteries from WD-fed mice were stiffer and less distensible, with marginally increased wall stress for a given strain, and a significantly increased Young's modulus of elasticity. Structurally, the wall cross-sectional area and the number of fenestrae found in the internal elastic lamina (IEL) of mesenteric arteries from mice fed a WD were significantly smaller than those of arteries from the ND-fed mice. There was also a significant increase in the volume of elastin, but not collagen in arteries from the WD cohort. Plasma levels of neuraminidase and the amount of TGF-β in mesenteric arteries were elevated in mice fed a WD, while ex vivo, cultured vascular smooth muscle cells exposed to neuraminidase secreted greater amounts of tropoelastin and TGF-β than those exposed to vehicle. These data suggest that consumption of a diet high in fat and sugar causes stiffening of the vascular wall in resistance arteries through a process that may involve increased neuraminidase and TGF-β activity, elevated production of elastin, and a reduction in the size and number of fenestrae in the arterial IEL.
Collapse
Affiliation(s)
| | - Jorge A. Castorena-Gonzalez
- Dalton Cardiovascular Research Center, University of MissouriColumbia, MO, USA
- Department of Biological Engineering, University of MissouriColumbia, MO, USA
| | - Francisco I. Ramirez-Perez
- Dalton Cardiovascular Research Center, University of MissouriColumbia, MO, USA
- Department of Biological Engineering, University of MissouriColumbia, MO, USA
| | - Guanghong Jia
- Diabetes and Cardiovascular Research Center, University of MissouriColumbia, MO, USA
- Harry S. Truman Memorial Veterans HospitalColumbia, MO, USA
| | - Michael A. Hill
- Dalton Cardiovascular Research Center, University of MissouriColumbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of MissouriColumbia, MO, USA
| | | | - James R. Sowers
- Diabetes and Cardiovascular Research Center, University of MissouriColumbia, MO, USA
- Harry S. Truman Memorial Veterans HospitalColumbia, MO, USA
| | - Luis A. Martinez-Lemus
- Dalton Cardiovascular Research Center, University of MissouriColumbia, MO, USA
- Department of Biological Engineering, University of MissouriColumbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of MissouriColumbia, MO, USA
- *Correspondence: Luis A. Martinez-Lemus
| |
Collapse
|
52
|
Qa'aty N, Vincent M, Wang Y, Wang A, Mitts TF, Hinek A. Synthetic ligands of the elastin receptor induce elastogenesis in human dermal fibroblasts via activation of their IGF-1 receptors. J Dermatol Sci 2015; 80:175-85. [PMID: 26475432 DOI: 10.1016/j.jdermsci.2015.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 08/19/2015] [Accepted: 10/01/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND We have previously reported that a mixture of peptides obtained after chemical or enzymatic degradation of bovine elastin, induced new elastogenesis in human skin. OBJECTIVE Now, we investigated the elastogenic potential of synthetic peptides mimicking the elastin-derived, VGVAPG sequence, IGVAPG sequence that we found in the rice bran, and a similar peptide, VGVTAG that we identified in the IGF-1-binding protein-1 (IGFBP-1). RESULTS We now demonstrate that treatment with each of these xGVxxG peptides (recognizable by the anti-elastin antibody), up-regulated the levels of elastin-encoding mRNA, tropoelastin protein, and the deposition of new elastic fibers in cultures of human dermal fibroblasts and in cultured explants of human skin. Importantly, we found that such induction of new elastogenesis may involve two parallel signaling pathways triggered after activation of IGF-1 receptor. In the first one, the xGVxxG peptides interact with the cell surface elastin receptor, thereby causing the downstream activation of the c-Src kinase and a consequent cross-activation of the adjacent IGF-1R, even in the absence of its principal ligand. In the second pathway their hydrophobic association with the N-terminal domain (VGVTAG) of the serum-derived IGFBP-1 induces conformational changes of this IGF-1 chaperone allowing for the release of its cargo and a consequent ligand-specific phosphorylation of IGF-1R. CONCLUSION We present a novel, clinically relevant mechanism in which products of partial degradation of dermal elastin may stimulate production of new elastic fibers by dermal fibroblasts. Our findings particularly encourage the use of biologically safe synthetic xGVxxG peptides for regeneration of the injured or aged human skin.
Collapse
Affiliation(s)
- Nour Qa'aty
- Physiology & Experimental Medicine Program, Hospital for Sick Children, ON, Canada; Institute of Medical Science, University of Toronto, ON, Canada
| | - Matthew Vincent
- Physiology & Experimental Medicine Program, Hospital for Sick Children, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada
| | - Yanting Wang
- Physiology & Experimental Medicine Program, Hospital for Sick Children, ON, Canada
| | - Andrew Wang
- Physiology & Experimental Medicine Program, Hospital for Sick Children, ON, Canada
| | | | - Aleksander Hinek
- Physiology & Experimental Medicine Program, Hospital for Sick Children, ON, Canada; Institute of Medical Science, University of Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada.
| |
Collapse
|
53
|
Neves JDC, Rizzato VR, Fappi A, Garcia MM, Chadi G, van de Vlekkert D, d'Azzo A, Zanoteli E. Neuraminidase-1 mediates skeletal muscle regeneration. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1755-64. [PMID: 26001931 PMCID: PMC5617636 DOI: 10.1016/j.bbadis.2015.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 05/06/2015] [Accepted: 05/11/2015] [Indexed: 12/31/2022]
Abstract
Neuraminidase-1 (NEU1) is the sialidase responsible for the catabolism of sialoglycoconjugates in lysosomes. Congenital NEU1 deficiency causes sialidosis, a severe lysosomal storage disease associated with a broad spectrum of clinical manifestations, which also include skeletal deformities, skeletal muscle hypotonia and weakness. Neu1(-/-) mice, a model of sialidosis, develop an atypical form of muscle degeneration caused by progressive expansion of the connective tissue that infiltrates the muscle bed, leading to fiber degeneration and atrophy. Here we investigated the role of Neu1 in the myogenic process that ensues during muscle regeneration after cardiotoxin-induced injury of limb muscles. A comparative analysis of cardiotoxin-treated muscles from Neu1(-/-) mice and Neu1(+/+) mice showed increased inflammatory and proliferative responses in the absence of Neu1 during the early stages of muscle regeneration. This was accompanied by significant and sequential upregulation of Pax7, MyoD, and myogenin mRNAs. The levels of both MyoD and myogenin proteins decreased during the late stages of regeneration, which most likely reflected an increased rate of degradation of the myogenic factors in the Neu1(-/-) muscle. We also observed a delay in muscle cell differentiation, which was characterized by prolonged expression of embryonic myosin heavy chain, as well as reduced myofiber cross-sectional area. At the end of the regenerative process, collagen type III deposition was increased compared to wild-type muscles and internal controls, indicating the initiation of fibrosis. Overall, these results point to a role of Neu1 throughout muscle regeneration.
Collapse
Affiliation(s)
| | | | - Alan Fappi
- Department of Neurology, University of São Paulo, São Paulo, SP 01246-903, Brazil
| | | | - Gerson Chadi
- Department of Neurology, University of São Paulo, São Paulo, SP 01246-903, Brazil
| | | | - Alessandra d'Azzo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Edmar Zanoteli
- Department of Neurology, University of São Paulo, São Paulo, SP 01246-903, Brazil.
| |
Collapse
|
54
|
Mecham RP, Gibson MA. The microfibril-associated glycoproteins (MAGPs) and the microfibrillar niche. Matrix Biol 2015; 47:13-33. [PMID: 25963142 DOI: 10.1016/j.matbio.2015.05.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/23/2015] [Accepted: 03/24/2015] [Indexed: 10/23/2022]
Abstract
The microfibril-associated glycoproteins MAGP-1 and MAGP-2 are extracellular matrix proteins that interact with fibrillin to influence microfibril function. The two proteins are related through a 60 amino acid matrix-binding domain but their sequences differ outside of this region. A distinguishing feature of both proteins is their ability to interact with TGFβ family growth factors, Notch and Notch ligands, and multiple elastic fiber proteins. MAGP-2 can also interact with αvβ3 integrins via a RGD sequence that is not found in MAGP-1. Morpholino knockdown of MAGP-1 expression in zebrafish resulted in abnormal vessel wall architecture and altered vascular network formation. In the mouse, MAGP-1 deficiency had little effect on elastic fibers in blood vessels and lung but resulted in numerous unexpected phenotypes including bone abnormalities, hematopoietic changes, increased fat deposition, diabetes, impaired wound repair, and a bleeding diathesis. Inactivation of the gene for MAGP-2 in mice produced a neutropenia yet had minimal effects on bone or adipose homeostasis. Double knockouts had phenotypes characteristic of each individual knockout as well as several additional traits only seen when both genes are inactivated. A common mechanism underlying all of the traits associated with the knockout phenotypes is altered TGFβ signaling. This review summarizes our current understanding of the function of the MAGPs and discusses ideas related to their role in growth factor regulation.
Collapse
Affiliation(s)
- Robert P Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Mark A Gibson
- School of Medical Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
55
|
Lu J, Isaji T, Im S, Fukuda T, Hashii N, Takakura D, Kawasaki N, Gu J. β-Galactoside α2,6-sialyltranferase 1 promotes transforming growth factor-β-mediated epithelial-mesenchymal transition. J Biol Chem 2014; 289:34627-41. [PMID: 25344606 DOI: 10.1074/jbc.m114.593392] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
β-Galactoside α2,6-sialyltranferase 1 (ST6GAL1) catalyzes the addition of terminal α2,6-sialylation to N-glycans. Increased expression of ST6GAL1 has been reported in diverse carcinomas and highly correlates with tumor progression. Here, we report that St6gal1 transcription and α2,6-sialylated N-glycans are up-regulated during TGF-β-induced epithelial-mesenchymal transition (EMT) in GE11 cells, requiring the Sp1 element within the St6gal1 promoter. Knockdown of St6gal1 strongly suppressed TGF-β-induced EMT with a concomitant increase in E-cadherin expression, a major determinant of epithelial cell adherens junctions. Conversely, overexpression of ST6GAL1 increased the turnover of cell surface E-cadherin and promoted TGF-β-induced EMT. Overexpressing β-galactoside α2,3-sialyltranferase 4 had little influence on EMT, indicating specificity for α2,6-sialylation. The basal mesenchymal phenotype of MDA-MB-231 human breast cancer cells was partially reversed by ST6GAL1 silencing. Moreover, ST6GAL1 knockdown inhibited the phosphorylation of Akt, but not Smad2, suggesting that ST6GAL1 contributes to EMT through a non-Smad signaling pathway. Taken together, our data indicate that ST6GAL1 promotes TGF-β-dependent EMT as well as maintenance of the mesenchymal state by growth signaling, providing a plausible mechanism whereby up-regulated ST6GAL1 may promote malignant progression.
Collapse
Affiliation(s)
- Jishun Lu
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558, Japan and
| | - Tomoya Isaji
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558, Japan and
| | - Sanghun Im
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558, Japan and
| | - Tomohiko Fukuda
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558, Japan and
| | - Noritaka Hashii
- the National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya, Tokyo, 158-8501, Japan
| | - Daisuke Takakura
- the National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya, Tokyo, 158-8501, Japan
| | - Nana Kawasaki
- the National Institute of Health Sciences, 1-18-1, Kamiyoga, Setagaya, Tokyo, 158-8501, Japan
| | - Jianguo Gu
- From the Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558, Japan and
| |
Collapse
|
56
|
Shiozaki K, Ryuzono S, Matsushita N, Ikeda A, Takeshita K, Chigwechokha PK, Komatsu M, Miyagi T. Molecular cloning and biochemical characterization of medaka (Oryzias latipes) lysosomal neu4 sialidase. FISH PHYSIOLOGY AND BIOCHEMISTRY 2014; 40:1461-1472. [PMID: 24744226 DOI: 10.1007/s10695-014-9940-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 04/09/2014] [Indexed: 06/03/2023]
Abstract
Glycoconjugates are known to be involved in many physiological events in vertebrates. Sialidase is one of the glycosidases, which removes sialic acid from glycoconjugates. In mammals, the properties and physiological functions of sialidases have been investigated, while there is little understanding of fish sialidase. Here, to investigate the significance of fish neu4 sialidase, neu4 gene was cloned from medaka brain mRNA and identified. Sialidase-specific motifs (GPG, YRVP and Asp-Box) were well conserved in the medaka neu4 polypeptide. Optimal pH of medaka neu4 sialidase was 4.6, but its activity was sustained even at neutral and weak alkaline pH. The neu4 considerably cleaved sialic acid from 4-methylumbelliferyl-N-acetyl-α-D-neuraminic acid and sialyllactose, but not from ganglioside and fetuin, which are good substrates for human NEU4. neu4 activity was mostly detected in mitochondria/lysosome fraction after biochemical fractionation, and indirect immunofluorescence assays revealed neu4 localization in lysosome in neu4 overexpressed cells. Next, developmental change in medaka neu4 and other sialidase mRNA levels were estimated by real-time PCR. Each sialidases showed different expression patterns in embryonic development: neu4 was up-regulated at late developmental stage in embryo, and neu3a mRNA level was quite high in 0.5 dpf. On the other hand, neu3b expression was drastically increased after hatching, suggesting that each sialidase may play a different role in embryonic development.
Collapse
Affiliation(s)
- Kazuhiro Shiozaki
- Faculty of Fisheries, Kagoshima University, 4-50-20 Shimo-arata, Kagoshima, 890-0056, Japan,
| | | | | | | | | | | | | | | |
Collapse
|
57
|
Pshezhetsky AV, Ashmarina LI. Desialylation of surface receptors as a new dimension in cell signaling. BIOCHEMISTRY (MOSCOW) 2014; 78:736-45. [PMID: 24010837 DOI: 10.1134/s0006297913070067] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Terminal sialic acid residues are found in abundance in glycan chains of glycoproteins and glycolipids on the surface of all live cells forming an outer layer of the cell originally known as glycocalyx. Their presence affects the molecular properties and structure of glycoconjugates, modifying their function and interactions with other molecules. Consequently, the sialylation state of glycoproteins and glycolipids has been recognized as a critical factor modulating molecular recognitions inside the cell, between the cells, between the cells and the extracellular matrix, and between the cells and certain exogenous pathogens. Until recently sialyltransferases that catalyze transfer of sialic acid residues to the glycan chains in the process of their biosynthesis were thought to be mainly responsible for the creation and maintenance of a temporal and spatial diversity of sialylated moieties. However, the growing evidence suggests that in mammalian cells, at least equally important roles belong to sialidases/neuraminidases, which are located on the cell surface and in intracellular compartments, and may either initiate the catabolism of sialoglycoconjugates or just cleave their sialic acid residues, and thereby contribute to temporal changes in their structure and functions. The current review summarizes emerging data demonstrating that mammalian neuraminidase 1, well known for its lysosomal catabolic function, is also targeted to the cell surface and assumes the previously unrecognized role as a structural and functional modulator of cellular receptors.
Collapse
Affiliation(s)
- A V Pshezhetsky
- Department of Medical Genetics, CHU Sainte-Justine Research Center, Montreal, Qc, H3T1C5, Canada.
| | | |
Collapse
|
58
|
Lysosomal multienzyme complex: pros and cons of working together. Cell Mol Life Sci 2013; 71:2017-32. [PMID: 24337808 DOI: 10.1007/s00018-013-1538-3] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 11/29/2013] [Accepted: 12/02/2013] [Indexed: 12/31/2022]
Abstract
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.
Collapse
|
59
|
Miao M, Reichheld SE, Muiznieks LD, Huang Y, Keeley FW. Elastin Binding Protein and FKBP65 Modulate in Vitro Self-Assembly of Human Tropoelastin. Biochemistry 2013; 52:7731-41. [DOI: 10.1021/bi400760f] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Ming Miao
- Research
Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Sean E. Reichheld
- Research
Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Lisa D. Muiznieks
- Research
Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Yayi Huang
- Research
Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Fred W. Keeley
- Department
of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
60
|
An eight-segment swine influenza virus harboring H1 and H3 hemagglutinins is attenuated and protective against H1N1 and H3N2 subtypes in pigs. J Virol 2013; 87:10114-25. [PMID: 23843633 DOI: 10.1128/jvi.01348-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Swine influenza virus (SIV) infections continue to cause production losses in the agricultural industry in addition to being a human public health concern. The primary method of controlling SIV is through vaccination. The killed SIV vaccines currently in use must be closely matched to the challenge virus, and their protective efficacy is limited. Live attenuated influenza vaccines (LAIV) provide strong, long-lived cell-mediated and humoral immunity against different influenza virus subtypes with no need for antigen matching. Here we report the generation of a new potential LAIV, an eight-segment SIV harboring two different SIV hemagglutinins (HAs), H1 and H3, in the genetic background of H1N1 SIV. This mutant SIV was generated by fusing the H3 HA ectodomain from A/Swine/Texas/4199-2/98 (H3N2) to the cytoplasmic tail, transmembrane domain, and stalk region of neuraminidase (NA) from A/Swine/Saskatchewan/18789/02 (H1N1) SIV. While this H1-H3 chimeric SIV, when propagated in vitro in the presence of exogenous neuraminidase, showed kinetics and growth properties similar to those of the parental wild-type virus, in vivo it was highly attenuated in pigs, demonstrating a great potential for serving as a dual LAIV. Furthermore, vaccination with the H1-H3 virus elicited robust immune responses, which conferred complete protection against infections with both H1 and H3 SIV subtypes in pigs.
Collapse
|
61
|
Dridi L, Seyrantepe V, Fougerat A, Pan X, Bonneil É, Thibault P, Moreau A, Mitchell GA, Heveker N, Cairo CW, Issad T, Hinek A, Pshezhetsky AV. Positive regulation of insulin signaling by neuraminidase 1. Diabetes 2013; 62:2338-46. [PMID: 23520133 PMCID: PMC3712076 DOI: 10.2337/db12-1825] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Neuraminidases (sialidases) catalyze the removal of sialic acid residues from sialylated glycoconjugates. We now report that mammalian neuraminidase 1 (Neu1), in addition to its catabolic function in lysosomes, is transported to the cell surface where it is involved in the regulation of insulin signaling. Insulin binding to its receptor rapidly induces interaction of the receptor with Neu1, which hydrolyzes sialic acid residues in the glycan chains of the receptor and, consequently, induces its activation. Cells from sialidosis patients with a genetic deficiency of Neu1 show impairment of insulin-induced phosphorylation of downstream protein kinase AKT, and treatment of these cells with purified Neu1 restores signaling. Genetically modified mice with ∼10% of the normal Neu1 activity exposed to a high-fat diet develop hyperglycemia and insulin resistance twice as fast as their wild-type counterparts. Together, these studies identify Neu1 as a novel component of the signaling pathways of energy metabolism and glucose uptake.
Collapse
Affiliation(s)
- Larbi Dridi
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montréal, Montréal, Québec, Canada
| | - Volkan Seyrantepe
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montréal, Montréal, Québec, Canada
| | - Anne Fougerat
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montréal, Montréal, Québec, Canada
| | - Xuefang Pan
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montréal, Montréal, Québec, Canada
| | - Éric Bonneil
- Institute of Research in Immunology and Cancer, University of Montréal, Montréal, Québec, Canada
| | - Pierre Thibault
- Institute of Research in Immunology and Cancer, University of Montréal, Montréal, Québec, Canada
| | - Allain Moreau
- Department of Stomatology, Faculty of Dentistry, University of Montréal, Montréal, Québec, Canada
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Sainte-Justine University Hospital Research Center, University of Montréal, Montréal, Québec, Canada
- Department of Biochemistry, University of Montréal, Montréal, Québec, Canada
| | - Grant A. Mitchell
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montréal, Montréal, Québec, Canada
| | - Nikolaus Heveker
- Department of Biochemistry, University of Montréal, Montréal, Québec, Canada
| | - Christopher W. Cairo
- Alberta Glycomics Centre, Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Tarik Issad
- Institut Cochin, Université Paris Descartes, CNRS (UMR8104), Paris, France
- INSERM, U1016, Paris, France
| | - Alexander Hinek
- Physiology and Experimental Medicine Program, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Alexey V. Pshezhetsky
- Division of Medical Genetics, Sainte-Justine University Hospital Research Center, University of Montréal, Montréal, Québec, Canada
- Department of Biochemistry, University of Montréal, Montréal, Québec, Canada
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montréal, Québec, Canada
- Corresponding author: Alexey V. Pshezhetsky,
| |
Collapse
|
62
|
D'Avila F, Tringali C, Papini N, Anastasia L, Croci G, Massaccesi L, Monti E, Tettamanti G, Venerando B. Identification of lysosomal sialidase NEU1 and plasma membrane sialidase NEU3 in human erythrocytes. J Cell Biochem 2013; 114:204-11. [PMID: 22903576 DOI: 10.1002/jcb.24355] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 08/07/2012] [Indexed: 11/11/2022]
Abstract
The sialylation level of molecules, sialoglycoproteins and gangliosides, protruding from plasma membranes regulates multiple facets of erythrocyte function, from interaction with endothelium to cell lifespan. Our results demonstrate that: (a) Both sialidases NEU1 and NEU3 are present on erythrocyte plasma membrane; (b) NEU1 is kept on the plasma membrane in absence of the protective protein/cathepsin A (PPCA); (c) NEU1 and NEU3 are retained on the plasma membrane, as peripheral proteins, associated to the external leaflet and released by alkaline treatments; (d) NEU1 and NEU3 are segregated in Triton X-100 detergent-resistant membrane domains (DRMs); (e) NEU3 shows activity also at neutral pH; and (f) NEU1 and NEU3 are progressively lost during erythrocyte life. Interestingly, sialidase activity released from erythrocyte membranes after an alkaline treatment preserves its functionality and recognizes sialoglycoproteins and gangliosides. On the other hand, the weak anchorage of sialidases to the plasma membrane and their loss during erythrocyte life could be a tool to preserve the cellular sialic acid content in order to avoid the early ageing of erythrocyte and processes of cell aggregation in the capillaries.
Collapse
Affiliation(s)
- Francesca D'Avila
- Department of Medical Chemistry, Biochemistry and Biotechnology, University of Milan, F.lli Cervi 93, Segrate, Milan 20090, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Abstract
Underlying the dynamic regulation of tropoelastin expression and elastin formation in development and disease are transcriptional and post-transcriptional mechanisms that have been the focus of much research. Of particular importance is the cytokine-governed elastin regulatory axis in which the pro-elastogenic activities of transforming growth factor β-1 (TGFβ1) and insulin-like growth factor-I (IGF-I) are opposed by anti-elastogenic activities of basic fibroblast growth factor (bFGF/FGF-2), heparin-binding epidermal growth factor-like growth factor (HB-EGF), EGF, PDGF-BB, TGFα, tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β and noncanonical TGFβ1 signaling. A key mechanistic feature of the regulatory axis is that cytokines influence elastin formation through effects on the cell cycle involving control of cyclin-cyclin dependent kinase complexes and activation of the Ras/MEK/ERK signaling pathway. In this article we provide an overview of the major cytokines/growth factors that modulate elastogenesis and describe the underlying molecular mechanisms for their action on elastin production.
Collapse
Affiliation(s)
- Erin P Sproul
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | |
Collapse
|
64
|
Fanzani A, Zanola A, Faggi F, Papini N, Venerando B, Tettamanti G, Sampaolesi M, Monti E. Implications for the mammalian sialidases in the physiopathology of skeletal muscle. Skelet Muscle 2012; 2:23. [PMID: 23114189 PMCID: PMC3534598 DOI: 10.1186/2044-5040-2-23] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 10/02/2012] [Indexed: 12/11/2022] Open
Abstract
The family of mammalian sialidases is composed of four distinct versatile enzymes that remove negatively charged terminal sialic acid residues from gangliosides and glycoproteins in different subcellular areas and organelles, including lysosomes, cytosol, plasma membrane and mitochondria. In this review we summarize the growing body of data describing the important role of sialidases in skeletal muscle, a complex apparatus involved in numerous key functions and whose functional integrity can be affected by various conditions, such as aging, chronic diseases, cancer and neuromuscular disorders. In addition to supporting the proper catabolism of glycoconjugates, sialidases can affect different signaling pathways by desialylation of many receptors and modulation of ganglioside content in cell membranes, thus actively participating in myoblast proliferation, differentiation and hypertrophy, insulin responsiveness and skeletal muscle architecture.
Collapse
Affiliation(s)
- Alessandro Fanzani
- Department of Biomedical Sciences and Biotechnologies and Interuniversitary Institute of Myology (IIM), University of Brescia, Viale Europa 11, 25123, Brescia, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
65
|
Kaptan E, Bas SS, Inceli MS. Total sialic acid profile in regressing and remodelling organs during the metamorphosis of marsh frog (Pelophylax ridibundus Pallas 1771). Cell Biochem Funct 2012; 31:173-9. [PMID: 22972462 DOI: 10.1002/cbf.2872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 07/21/2012] [Accepted: 08/08/2012] [Indexed: 11/10/2022]
Abstract
This study aimed to investigate the functional relationship of sialic acid in regressing and remodelling organs such as the tail, small intestine and liver during the metamorphosis of Pelophylax ridibundus. For this purpose, four groups were composed according to developmental periods by considering Gosner's criteria (1964). Our findings showed that the sialic acid content of the larval tail has an opposite profile to cell death process. Although the sialic acid content of the small intestine and liver did not change evidently during metamorphosis, it increased after the completion of metamorphosis. Frog tail extensively exhibited cell death process and decreased proliferative activity and underwent complete degeneration during metamorphic climax. In spite of increased apoptotic index, a decreased sialic acid level in the tail tissues during climax can be the indication of a death cell removal process. However, the intestine and the liver included both cell death and proliferative process and remodelling in their adult forms. Thus, their sialic acid profiles during metamorphosis were different from the tail's profile. These data show that sialic acid may be an indicator of the presence of some cellular events during metamorphosis and that it can have different roles in the developmental process depending on the organ's fate throughout metamorphosis.
Collapse
Affiliation(s)
- Engin Kaptan
- Department of Biology, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Turkey.
| | | | | |
Collapse
|
66
|
Abstract
Sialic acids are a diverse family of monosaccharides widely expressed on all cell surfaces of vertebrates and so-called "higher" invertebrates, and on certain bacteria that interact with vertebrates. This overview surveys examples of biological roles of sialic acids in immunity, with emphasis on an evolutionary perspective. Given the breadth of the subject, the treatment of individual topics is brief. Subjects discussed include biophysical effects regulation of factor H; modulation of leukocyte trafficking via selectins; Siglecs in immune cell activation; sialic acids as ligands for microbes; impact of microbial and endogenous sialidases on immune cell responses; pathogen molecular mimicry of host sialic acids; Siglec recognition of sialylated pathogens; bacteriophage recognition of microbial sialic acids; polysialic acid modulation of immune cells; sialic acids as pathogen decoys or biological masks; modulation of immunity by sialic acid O-acetylation; sialic acids as antigens and xeno-autoantigens; antisialoglycan antibodies in reproductive incompatibility; and sialic-acid-based blood groups.
Collapse
Affiliation(s)
- Ajit Varki
- Glycobiology Research and Training Center, Department of Medicine, University of California at San Diego, La Jolla, 92093-0687, USA.
| | | |
Collapse
|
67
|
Miyagi T, Takahashi K, Hata K, Shiozaki K, Yamaguchi K. Sialidase significance for cancer progression. Glycoconj J 2012; 29:567-77. [PMID: 22644327 DOI: 10.1007/s10719-012-9394-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 05/05/2012] [Accepted: 05/08/2012] [Indexed: 11/26/2022]
Abstract
Aberrant glycosylation is a characteristic feature of cancer cells. In particular, altered sialylation is closely associated with malignant properties, including invasiveness and metastatic potential. To elucidate the molecular mechanisms underlying the aberrancy, our studies have focused on mammalian sialidase, which catalyzes the removal of sialic acid residues from glycoproteins and glycolipids. The four types of mammalian sialidase identified to date show altered expression and behave in different manners during carcinogenesis. The present review briefly summarizes results on altered expression of sialidases and their possible roles in cancer progression. These enzymes are indeed factors defining cancer malignancy and thus potential targets for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Taeko Miyagi
- Division of Cancer Glycosylation Research, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, Japan.
| | | | | | | | | |
Collapse
|
68
|
Feng C, Stamatos NM, Dragan AI, Medvedev A, Whitford M, Zhang L, Song C, Rallabhandi P, Cole L, Nhu QM, Vogel SN, Geddes CD, Cross AS. Sialyl residues modulate LPS-mediated signaling through the Toll-like receptor 4 complex. PLoS One 2012; 7:e32359. [PMID: 22496731 PMCID: PMC3322133 DOI: 10.1371/journal.pone.0032359] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 01/26/2012] [Indexed: 11/19/2022] Open
Abstract
We previously reported that neuraminidase (NA) pretreatment of human PBMCs markedly increased their cytokine response to lipopolysaccharide (LPS). To study the mechanisms by which this occurs, we transfected HEK293T cells with plasmids encoding TLR4, CD14, and MD2 (three components of the LPS receptor complex), as well as a NFκB luciferase reporting system. Both TLR4 and MD2 encoded by the plasmids are α-2,6 sialylated. HEK293T cells transfected with TLR4/MD2/CD14 responded robustly to the addition of LPS; however, omission of the MD2 plasmid abrogated this response. Addition of culture supernatants from MD2 (sMD2)-transfected HEK293T cells, but not recombinant, non-glycosylated MD2 reconstituted this response. NA treatment of sMD2 enhanced the LPS response as did NA treatment of the TLR4/CD14-transfected cell supplemented with untreated sMD2, but optimal LPS-initiated responses were observed with NA-treated TLR4/CD14-transfected cells supplemented with NA-treated sMD2. We hypothesized that removal of negatively charged sialyl residues from glycans on the TLR4 complex would hasten the dimerization of TLR4 monomers required for signaling. Co-transfection of HEK293T cells with separate plasmids encoding either YFP- or FLAG-tagged TLR4, followed by treatment with NA and stimulation with LPS, led to an earlier and more robust time-dependent dimerization of TLR4 monomers on co-immunoprecipitation, compared to untreated cells. These findings were confirmed by fluorescence resonance energy transfer (FRET) analysis. Overexpression of human Neu1 increased LPS-initiated TLR4-mediated NFκB activation and a NA inhibitor suppressed its activation. We conclude that (1) sialyl residues on TLR4 modulate LPS responsiveness, perhaps by facilitating clustering of the homodimers, and that (2) sialic acid, and perhaps other glycosyl species, regulate MD2 activity required for LPS-mediated signaling. We speculate that endogenous sialidase activity mobilized during cell activation may play a role in this regulation.
Collapse
Affiliation(s)
- Chiguang Feng
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Nicholas M. Stamatos
- Institute of Human Virology and Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Anatoliy I. Dragan
- Institute of Fluorescence, University of Maryland, Baltimore County, Baltimore, Maryland, United States of America
| | - Andrei Medvedev
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Melissa Whitford
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Lei Zhang
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Chang Song
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Prasad Rallabhandi
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Leah Cole
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Quan M. Nhu
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Stefanie N. Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Chris D. Geddes
- Institute of Fluorescence, University of Maryland, Baltimore County, Baltimore, Maryland, United States of America
| | - Alan S. Cross
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
69
|
Kościelak J. The hypothesis on function of glycosphingolipids and ABO blood groups revisited. Neurochem Res 2012; 37:1170-84. [PMID: 22407244 DOI: 10.1007/s11064-012-0734-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Revised: 12/23/2011] [Accepted: 02/15/2012] [Indexed: 12/12/2022]
Abstract
Twenty-five years ago the author proposed new ideas of glycoprotein (GPs) and glycosphingolipid (GSLs) functions at the cell membrane. The GPs, apart from their glycan carrying capacity, were assumed to have specific, protein associated, functions. In contrast, GSLs such as those of globo and neolacto/lacto series, were considered to be energetically cheap membrane packing substances, filling in membrane spaces not covered with functional GPs. The terminal carbohydrate structures of the neolacto/lacto GSLs, i.e., sialic acid residues and ABH glycotopes, were postulated to have either regulatory or protective functions, respectively. A special active role was ascribed to terminal β-galactosyl residues of GSLs and GPs. Gangliosides were considered to be functional GSLs. In the present review the author discusses these old ideas in context of the contemporary knowledge and comes to the conclusion that they have not aged.
Collapse
Affiliation(s)
- Jerzy Kościelak
- Department of Biochemistry, Institute of Hematology and Transfusion Medicine, Warsaw, Poland.
| |
Collapse
|
70
|
Cross AS, Hyun SW, Miranda-Ribera A, Feng C, Liu A, Nguyen C, Zhang L, Luzina IG, Atamas SP, Twaddell WS, Guang W, Lillehoj EP, Puché AC, Huang W, Wang LX, Passaniti A, Goldblum SE. NEU1 and NEU3 sialidase activity expressed in human lung microvascular endothelia: NEU1 restrains endothelial cell migration, whereas NEU3 does not. J Biol Chem 2012; 287:15966-80. [PMID: 22403397 DOI: 10.1074/jbc.m112.346817] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The microvascular endothelial surface expresses multiple molecules whose sialylation state regulates multiple aspects of endothelial function. To better regulate these sialoproteins, we asked whether endothelial cells (ECs) might express one or more catalytically active sialidases. Human lung microvascular EC lysates contained heat-labile sialidase activity for a fluorogenic substrate, 2'-(4-methylumbelliferyl)-α-D-N-acetylneuraminic acid (4-MU-NANA), that was dose-dependently inhibited by the competitive sialidase inhibitor, 2,3-dehydro-2-deoxy-N-acetylneuraminic acid but not its negative control. The EC lysates also contained sialidase activity for a ganglioside mixture. Using real time RT-PCR to detect mRNAs for the four known mammalian sialidases, NEU1, -2, -3, and -4, NEU1 mRNA was expressed at levels 2700-fold higher that those found for NEU2, -3, or -4. Western analyses indicated NEU1 and -3 protein expression. Using confocal microscopy and flow cytometry, NEU1 was immunolocalized to both the plasma membrane and the perinuclear region. NEU3 was detected both in the cytosol and nucleus. Prior siRNA-mediated knockdown of NEU1 and NEU3 each decreased EC sialidase activity for 4-MU-NANA by >65 and >17%, respectively, and for the ganglioside mixture by 0 and 40%, respectively. NEU1 overexpression in ECs reduced their migration into a wound by >40%, whereas NEU3 overexpression did not. Immunohistochemical studies of normal human tissues immunolocalized NEU1 and NEU3 proteins to both pulmonary and extrapulmonary vascular endothelia. These combined data indicate that human lung microvascular ECs as well as other endothelia express catalytically active NEU1 and NEU3. NEU1 restrains EC migration, whereas NEU3 does not.
Collapse
Affiliation(s)
- Alan S Cross
- Center for Vaccine Development, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Takahashi K, Mitoma J, Hosono M, Shiozaki K, Sato C, Yamaguchi K, Kitajima K, Higashi H, Nitta K, Shima H, Miyagi T. Sialidase NEU4 hydrolyzes polysialic acids of neural cell adhesion molecules and negatively regulates neurite formation by hippocampal neurons. J Biol Chem 2012; 287:14816-26. [PMID: 22393058 DOI: 10.1074/jbc.m111.324186] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Modulation of levels of polysialic acid (polySia), a sialic acid polymer, predominantly associated with the neural cell adhesion molecule (NCAM), influences neural functions, including synaptic plasticity, neurite growth, and cell migration. Biosynthesis of polySia depends on two polysialyltransferases ST8SiaII and ST8SiaIV in vertebrate. However, the enzyme involved in degradation of polySia in its physiological turnover remains uncertain. In the present study, we identified and characterized a murine sialidase NEU4 that catalytically degrades polySia. Murine NEU4, dominantly expressed in the brain, was found to efficiently hydrolyze oligoSia and polySia chains as substrates in sialidase in vitro assays, and also NCAM-Fc chimera as well as endogenous NCAM in tissue homogenates of postnatal mouse brain as assessed by immunoblotting with anti-polySia antibodies. Degradation of polySia by NEU4 was also evident in neuroblastoma Neuro2a cells that were co-transfected with Neu4 and ST8SiaIV genes. Furthermore, in mouse embryonic hippocampal primary neurons, the endogenously expressed NEU4 was found to decrease during the neuronal differentiation. Interestingly, GFP- or FLAG-tagged NEU4 was partially co-localized with polySia in neurites and significantly suppressed their outgrowth, whereas silencing of NEU4 showed the acceleration together with an increase in polySia expression. These results suggest that NEU4 is involved in regulation of neuronal function by polySia degradation in mammals.
Collapse
Affiliation(s)
- Kohta Takahashi
- Division of Cancer Glycosylation Research, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, 981-8558, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Miyagi T, Yamaguchi K. Mammalian sialidases: physiological and pathological roles in cellular functions. Glycobiology 2012; 22:880-96. [PMID: 22377912 DOI: 10.1093/glycob/cws057] [Citation(s) in RCA: 269] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Sialic acids are terminal acidic monosaccharides, which influence the chemical and biological features of glycoconjugates. Their removal catalyzed by a sialidase modulates various biological processes through change in conformation and creation or loss of binding sites of functional molecules. Sialidases exist widely in vertebrates and also in a variety of microorganisms. Recent research on mammalian sialidases has provided evidence for great importance of these enzymes in various cellular functions, including lysosomal catabolism, whereas microbial sialidases appear to play roles limited to nutrition and pathogenesis. Four types of mammalian sialidases have been identified and characterized to date, designated as NEU1, NEU2, NEU3 and NEU4. They are encoded by different genes and differ in major subcellular localization and enzymatic properties including substrate specificity, and each has been found to play a unique role depending on its particular properties. This review is an attempt to concisely summarize current knowledge concerning mammalian sialidases, with a special focus on their properties and physiological and pathological roles in cellular functions.
Collapse
Affiliation(s)
- Taeko Miyagi
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, Japan.
| | | |
Collapse
|
73
|
Lillehoj EP, Hyun SW, Feng C, Zhang L, Liu A, Guang W, Nguyen C, Luzina IG, Atamas SP, Passaniti A, Twaddell WS, Puché AC, Wang LX, Cross AS, Goldblum SE. NEU1 sialidase expressed in human airway epithelia regulates epidermal growth factor receptor (EGFR) and MUC1 protein signaling. J Biol Chem 2012; 287:8214-31. [PMID: 22247545 DOI: 10.1074/jbc.m111.292888] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Epithelial cells (ECs) lining the airways provide a protective barrier between the external environment and the internal host milieu. These same airway epithelia express receptors that respond to danger signals and initiate repair programs. Because the sialylation state of a receptor can influence its function and is dictated in part by sialidase activity, we asked whether airway epithelia express catalytically active sialidase(s). Human primary small airway and A549 ECs expressed NEU1 sialidase at the mRNA and protein levels, and NEU1 accounted for >70% of EC sialidase activity. Blotting with Maackia amurensis and peanut agglutinin lectins established epidermal growth factor receptor (EGFR) and MUC1 as in vivo substrates for NEU1. NEU1 associated with EGFR and MUC1, and NEU1-EGFR association was regulated by EGF stimulation. NEU1 overexpression diminished EGF-stimulated EGFR Tyr-1068 autophosphorylation by up to 44% but enhanced MUC1-dependent Pseudomonas aeruginosa adhesion by 1.6-1.7-fold and flagellin-stimulated ERK1/2 activation by 1.7-1.9-fold. In contrast, NEU1 depletion increased EGFR activation (1.5-fold) and diminished MUC1-mediated bacterial adhesion (38-56%) and signaling (73%). These data indicate for the first time that human airway epithelia express catalytically active NEU1 sialidase that regulates EGFR- and MUC1-dependent signaling and bacterial adhesion. NEU1 catalytic activity may offer an additional level of regulation over the airway epithelial response to ligands, pathogens, and injurious stimuli.
Collapse
Affiliation(s)
- Erik P Lillehoj
- Departments of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Abstract
The removal of sialic acids, catalyzed by sialidase, is the initial step in degradation of oligosaccharides, glycoproteins, and glycolipids. The catalytic reaction may greatly influence biological processes through changing the conformation of glycoproteins and create or mask binding sites of functional molecules. Recent progress in sialidase research has clarified that mammalian sialidases indeed contribute to the regulation of various cellular functions as well as lysosomal catabolism, unlike the sialidases of microbial origin that probably play roles limited to nutrition and pathogenesis. However, the mammalian enzymes contain consensus sequences in the six-blade β-propeller structural organization typical of microbial sialidases, despite the low degree of similarity to the amino acid sequences of the microbial enzymes. The present review briefly summarizes structural and functional features of mammalian sialidases.
Collapse
Affiliation(s)
- Eugenio Monti
- Faculty of Medicine, Department of Biomedical Sciences and Biotechnology, University of Brescia, Brescia, Viale Europa 11, 25123, Brescia, Italy
| | | |
Collapse
|
75
|
Where catabolism meets signalling: neuraminidase 1 as a modulator of cell receptors. Glycoconj J 2011; 28:441-52. [PMID: 21928149 DOI: 10.1007/s10719-011-9350-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 02/24/2011] [Accepted: 03/08/2011] [Indexed: 12/19/2022]
Abstract
Terminal sialic acid residues are found in abundance in glycan chains of glycoproteins and glycolipids on the surface of all live cells forming an outer layer of the cell originally known as glycocalyx. Their presence affects the molecular properties and structure of glycoconjugates, modifying their function and interactions with other molecules. Consequently, the sialylation state of glycoproteins and glycolipids has been recognized as a critical factor modulating molecular recognitions inside the cell, between the cells, between the cells and the extracellular matrix, and between the cells and certain exogenous pathogens. Sialyltransferases that attach sialic acid residues to the glycan chains in the process of their initial synthesis were thought to be mainly responsible for the creation and maintenance of a temporal and spatial diversity of sialylated moieties. However, the growing evidence also suggests that in mammalian cells, at least equally important roles belong to sialidases/neuraminidases, which are located on the cell surface and in intracellular compartments, and may either initiate the catabolism of sialoglycoconjugates or just cleave their sialic acid residues, and thereby contribute to temporal changes in their structure and functions. The current review summarizes emerging data demonstrating that neuraminidase 1 (NEU1), well known for its lysosomal catabolic function, can be also targeted to the cell surface and assume the previously unrecognized role as a structural and functional modulator of cellular receptors.
Collapse
|
76
|
Stamatos NM, Carubelli I, van de Vlekkert D, Bonten EJ, Papini N, Feng C, Venerando B, d'Azzo A, Cross AS, Wang LX, Gomatos PJ. LPS-induced cytokine production in human dendritic cells is regulated by sialidase activity. J Leukoc Biol 2010; 88:1227-39. [PMID: 20826611 DOI: 10.1189/jlb.1209776] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Nicholas M Stamatos
- University of Maryland Medical Center, 725 West Lombard St., Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Muscle degeneration in neuraminidase 1-deficient mice results from infiltration of the muscle fibers by expanded connective tissue. Biochim Biophys Acta Mol Basis Dis 2010; 1802:659-72. [PMID: 20388541 DOI: 10.1016/j.bbadis.2010.04.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 03/31/2010] [Accepted: 04/05/2010] [Indexed: 11/22/2022]
Abstract
Neuraminidase 1 (NEU1) regulates the catabolism of sialoglycoconjugates in lysosomes. Congenital NEU1 deficiency in children is the basis of sialidosis, a severe neurosomatic disorder in which patients experience a broad spectrum of clinical manifestations varying in the age of onset and severity. Osteoskeletal deformities and muscle hypotonia have been described in patients with sialidosis. Here we present the first comprehensive analysis of the skeletal muscle pathology associated with loss of Neu1 function in mice. In this animal model, skeletal muscles showed an expansion of the epimysial and perimysial spaces, associated with proliferation of fibroblast-like cells and abnormal deposition of collagens. Muscle fibers located adjacent to the expanded connective tissue underwent extensive invagination of their sarcolemma, which resulted in the infiltration of the fibers by fibroblast-like cells and extracellular matrix, and in their progressive cytosolic fragmentation. Both the expanded connective tissue and the juxtaposed infiltrated muscle fibers were strongly positive for lysosomal markers and displayed increased proteolytic activity of lysosomal cathepsins and metalloproteinases. These combined features could lead to abnormal remodeling of the extracellular matrix that could be responsible for sarcolemmal invagination and progressive muscle fiber degeneration, ultimately resulting in an overt atrophic phenotype. This unique pattern of muscle damage, which has never been described in any myopathy, might explain the neuromuscular manifestations reported in patients with the type II severe form of sialidosis. More broadly, these findings point to a potential role of NEU1 in cell proliferation and extracellular matrix remodeling.
Collapse
|
78
|
Arabkhari M, Bunda S, Wang Y, Wang A, Pshezhetsky AV, Hinek A. Desialylation of insulin receptors and IGF-1 receptors by neuraminidase-1 controls the net proliferative response of L6 myoblasts to insulin. Glycobiology 2010; 20:603-16. [PMID: 20100694 DOI: 10.1093/glycob/cwq010] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We recently established that the subunit of cell surface-residing elastin receptor, neuraminidase-1 (Neu1), can desialylate adjacent insulin-like growth factor 1 receptors (IGF-1R) of arterial smooth muscle cells, thereby quenching their proliferative response to insulin-like growth factor II. In this study, we explored whether Neu1 would also desialylate the insulin receptors (IR), as well as the IGF-1R on rat skeletal L6 myoblasts, and whether desialylation of IR and IGF-1R would affect a net proliferative effect of insulin. First, we found that physiological (0.5-1 nM) and high therapeutic (10 nM) insulin concentrations induced a modest increase in proliferation rate of cultured L6 myoblasts. While IR kinase inhibitor could abolish the mitogenic effect of these insulin concentrations, the observed more pronounced proliferative response to supraphysiological concentration (100 nM) of insulin could be eliminated only by specific inhibition of IGF-1R. Then, we found that treatment of L6 cells with mouse-derived Neu1 or with Clostridium perfringens neuraminidase caused desialylation of IR, which coincided with a significant increase of their proliferative response to lower (0.5-10 nM) concentrations of insulin. In contrast, experimental desialylation of IGF-1R coincided with elimination of the heightened proliferative response of L6 myoblasts to 100 nM insulin. Importantly, we also found that inhibition of endogenous Neu1 abolished the increase in proliferation of L6 cells induced by 1 and 10 nM of insulin, but amplified the proliferative effect of 100 nM insulin. We therefore conclude that desialylation of both IR and IGF-1R by Neu1 controls the net proliferative response of skeletal myoblasts to insulin.
Collapse
Affiliation(s)
- Majid Arabkhari
- Physiology and Experimental Medicine Program, The Hospital for Sick Children, University of Toronto, Canada
| | | | | | | | | | | |
Collapse
|
79
|
Monti E, Bonten; E, D'Azzo A, Bresciani R, Venerando B, Borsani G, Schauer R, Tettamanti G. Sialidases in Vertebrates. Adv Carbohydr Chem Biochem 2010; 64:403-79. [DOI: 10.1016/s0065-2318(10)64007-3] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
80
|
Seyrantepe V, Iannello A, Liang F, Kanshin E, Jayanth P, Samarani S, Szewczuk MR, Ahmad A, Pshezhetsky AV. Regulation of phagocytosis in macrophages by neuraminidase 1. J Biol Chem 2009; 285:206-15. [PMID: 19889639 DOI: 10.1074/jbc.m109.055475] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The differentiation of monocytes into macrophages and dendritic cells is accompanied by induction of cell-surface neuraminidase 1 (Neu1) and cathepsin A (CathA), the latter forming a complex with and activating Neu1. To clarify the biological importance of this phenomenon we have developed the gene-targeted mouse models of a CathA deficiency (CathA(S190A)) and a double CathA/Neu1 deficiency (CathA(S190A-Neo)). Macrophages of CathA(S190A-Neo) mice and their immature dendritic cells showed a significantly reduced capacity to engulf Gram-positive and Gram-negative bacteria and positively and negatively charged polymer beads as well as IgG-opsonized beads and erythrocytes. Properties of the cells derived from CathA(S190A) mice were indistinguishable from those of wild-type controls, suggesting that the absence of Neu1, which results in the increased sialylation of the cell surface proteins, probably affects multiple receptors for phagocytosis. Indeed, treatment of the cells with purified mouse Neu1 reduced surface sialylation and restored phagocytosis. Because Neu1-deficient cells showed reduced internalization of IgG-opsonized sheep erythrocytes whereas binding of the erythrocytes to the cells at 4 degrees C persisted, we speculate that the absence of Neu1 in particular affected transduction of signals from the Fc receptors for immunoglobulin G (FcgammaR). Indeed the macrophages from the Neu1-deficient mice showed increased sialylation and impaired phosphorylation of FcgammaR as well as markedly reduced phosphorylation of Syk kinase in response to treatment with IgG-opsonized beads. Altogether our data suggest that the cell surface Neu1 activates the phagocytosis in macrophages and dendritic cells through desialylation of surface receptors, thus, contributing to their functional integrity.
Collapse
Affiliation(s)
- Volkan Seyrantepe
- Sainte-Justine University Hospital Centre, University of Montreal, Montreal, QuebecH3T 1C5, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|