Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Proc Natl Acad Sci U S A ; 121(6): e2314309121, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38285943

ABSTRACT

Mucins are large, highly glycosylated extracellular matrix proteins that line and protect epithelia of the respiratory, digestive, and urogenital tracts. Previous work has shown that mucins form large, interconnected polymeric networks that mediate their biological functions once secreted. However, how these large matrix molecules are compacted and packaged into much smaller secretory granules within cells prior to secretion is largely unknown. Here, we demonstrate that a small cysteine-rich adaptor protein is essential for proper packaging of a secretory mucin in vivo. This adaptor acts via cysteine bonding between itself and the cysteine-rich domain of the mucin. Loss of this adaptor protein disrupts mucin packaging in secretory granules, alters the mobile fraction within granules, and results in granules that are larger, more circular, and more fragile. Understanding the factors and mechanisms by which mucins and other highly glycosylated matrix proteins are properly packaged and secreted may provide insight into diseases characterized by aberrant mucin secretion.


Subject(s)
Cysteine , Mucins , Mucins/metabolism , Cysteine/metabolism , Biological Transport , Secretory Vesicles/metabolism
2.
Proc Natl Acad Sci U S A ; 120(43): e2303703120, 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37862385

ABSTRACT

The family of GalNAc-Ts (GalNAcpolypeptide:N-Acetylgalactosaminyl transferases) catalyzes the first committed step in the synthesis of O-glycans, which is an abundant and biologically important protein modification. Abnormalities in the activity of individual GalNAc-Ts can result in congenital disorders of O-glycosylation (CDG) and influence a broad array of biological functions. How site-specific O-glycans regulate biology is unclear. Compiling in vivo O-glycosites would be an invaluable step in determining the function of site-specific O-glycans. We integrated chemical and enzymatic conditions that cleave O-glycosites, a higher-energy dissociation product ions-triggered electron-transfer/higher-energy collision dissociation mass spectrometry (MS) workflow and software to study nine mouse tissues and whole blood. We identified 2,154 O-glycosites from 595 glycoproteins. The O-glycosites and glycoproteins displayed consensus motifs and shared functions as classified by Gene Ontology terms. Limited overlap of O-glycosites was observed with protein O-GlcNAcylation and phosphorylation sites. Quantitative glycoproteomics and proteomics revealed a tissue-specific regulation of O-glycosites that the differential expression of Galnt isoenzymes in tissues partly contributes to. We examined the Galnt2-null mouse model, which phenocopies congenital disorder of glycosylation involving GALNT2 and revealed a network of glycoproteins that lack GalNAc-T2-specific O-glycans. The known direct and indirect functions of these glycoproteins appear consistent with the complex metabolic phenotypes observed in the Galnt2-null animals. Through this study and interrogation of databases and the literature, we have compiled an atlas of experimentally identified mouse O-glycosites consisting of 2,925 O-glycosites from 758 glycoproteins.


Subject(s)
Glycoproteins , Metabolic Diseases , Animals , Mice , Glycosylation , Glycoproteins/genetics , Glycoproteins/metabolism , Proteome/metabolism , Polysaccharides , Polypeptide N-acetylgalactosaminyltransferase
3.
J Biol Chem ; 300(4): 107164, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38484798

ABSTRACT

O-glycosylation is a conserved posttranslational modification that impacts many aspects of organismal viability and function. Recent studies examining the glycosyltransferase Galnt11 demonstrated that it glycosylates the endocytic receptor megalin in the kidneys, enabling proper binding and reabsorption of ligands, including vitamin D-binding protein (DBP). Galnt11-deficient mice were unable to properly reabsorb DBP from the urine. Vitamin D plays an essential role in mineral homeostasis and its deficiency is associated with bone diseases such as rickets, osteomalacia, and osteoporosis. We therefore set out to examine the effects of the loss of Galnt11 on vitamin D homeostasis and bone composition. We found significantly decreased levels of serum 25-hydroxyvitamin D and 1,25-dihydroxyvitamin D, consistent with decreased reabsorption of DBP. This was accompanied by a significant reduction in blood calcium levels and a physiologic increase in parathyroid hormone (PTH) in Galnt11-deficient mice. Bones in Galnt11-deficient mice were smaller and displayed a decrease in cortical bone accompanied by an increase in trabecular bone and an increase in a marker of bone formation, consistent with PTH-mediated effects on bone. These results support a unified model for the role of Galnt11 in bone and mineral homeostasis, wherein loss of Galnt11 leads to decreased reabsorption of DBP by megalin, resulting in a cascade of disrupted mineral and bone homeostasis including decreased circulating vitamin D and calcium levels, a physiological increase in PTH, an overall loss of cortical bone, and an increase in trabecular bone. Our study elucidates how defects in O-glycosylation can influence vitamin D and mineral homeostasis and the integrity of the skeletal system.


Subject(s)
Bone and Bones , Homeostasis , Polypeptide N-acetylgalactosaminyltransferase , Vitamin D , Animals , Male , Mice , Bone and Bones/anatomy & histology , Bone and Bones/chemistry , Bone and Bones/metabolism , Calcium/metabolism , Glycosylation , Homeostasis/genetics , Parathyroid Hormone/metabolism , Vitamin D/metabolism , Vitamin D/analogs & derivatives , Vitamin D-Binding Protein/metabolism
4.
Proc Natl Acad Sci U S A ; 119(43): e2209750119, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36252017

ABSTRACT

Mucins are large, highly glycosylated transmembrane and secreted proteins that line and protect epithelial surfaces. However, the details of mucin biosynthesis and packaging in vivo are largely unknown. Here, we demonstrate that multiple distinct mucins undergo intragranular restructuring during secretory granule maturation in vivo, forming unique structures that are spatially segregated within the same granule. We further identify temporally-regulated genes that influence mucin restructuring, including those controlling pH (Vha16-1), Ca2+ ions (fwe) and Cl- ions (Clic and ClC-c). Finally, we show that altered mucin glycosylation influences the dimensions of these structures, thereby affecting secretory granule morphology. This study elucidates key steps and factors involved in intragranular, rather than intergranular segregation of mucins through regulated restructuring events during secretory granule maturation. Understanding how multiple distinct mucins are efficiently packaged into and secreted from secretory granules may provide insight into diseases resulting from defects in mucin secretion.


Subject(s)
Mucins , Secretory Vesicles , Cytoplasmic Granules/metabolism , Glycosylation , Mucins/metabolism , Secretory Vesicles/metabolism
5.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34732583

ABSTRACT

The SARS-CoV-2 coronavirus responsible for the global pandemic contains a novel furin cleavage site in the spike protein (S) that increases viral infectivity and syncytia formation in cells. Here, we show that O-glycosylation near the furin cleavage site is mediated by members of the GALNT enzyme family, resulting in decreased furin cleavage and decreased syncytia formation. Moreover, we show that O-glycosylation is dependent on the novel proline at position 681 (P681). Mutations of P681 seen in the highly transmissible alpha and delta variants abrogate O-glycosylation, increase furin cleavage, and increase syncytia formation. Finally, we show that GALNT family members capable of glycosylating S are expressed in human respiratory cells that are targets for SARS-CoV-2 infection. Our results suggest that host O-glycosylation may influence viral infectivity/tropism by modulating furin cleavage of S and provide mechanistic insight into the role of the P681 mutations found in the highly transmissible alpha and delta variants.


Subject(s)
SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Animals , Cell Fusion , Cell Line , Furin/metabolism , Giant Cells , Glycosylation , Humans , N-Acetylgalactosaminyltransferases/metabolism , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Polypeptide N-acetylgalactosaminyltransferase
6.
Glycobiology ; 33(6): 476-489, 2023 06 21.
Article in English | MEDLINE | ID: mdl-37115803

ABSTRACT

The COVID-19 global pandemic has underscored the need to understand how viruses and other pathogens are able to infect and replicate within the respiratory system. Recent studies have highlighted the role of highly O-glycosylated mucins in the protection of the respiratory system as well as how mucin-type O-glycosylation may be able to modify viral infectivity. Therefore, we set out to identify the specific genes controlling mucin-type O-glycosylation throughout the mouse respiratory system as well as determine how their expression and the expression of respiratory mucins is influenced by infection or injury. Here, we show that certain mucins and members of the Galnt family are abundantly expressed in specific respiratory tissues/cells and demonstrate unique patterns of O-glycosylation across diverse respiratory tissues. Moreover, we find that the expression of certain Galnts and mucins is altered during lung infection and injury in experimental mice challenged with infectious agents, toxins, and allergens. Finally, we examine gene expression changes of Galnts and mucins in a mouse model of SARS-CoV-2 infection. Our work provides foundational knowledge regarding the specific expression of Galnt enzyme family members and mucins throughout the respiratory system, and how their expression is altered upon lung infection and injury.


Subject(s)
COVID-19 , Mucins , Animals , Mice , Mucins/genetics , Mucins/metabolism , Glycosylation , COVID-19/genetics , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Respiratory System/metabolism
7.
Proc Natl Acad Sci U S A ; 116(50): 25196-25202, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31740596

ABSTRACT

Chronic kidney disease (CKD) affects more than 20 million Americans and ∼10% of the population worldwide. Genome-wide association studies (GWAS) of kidney functional decline have identified genes associated with CKD, but the precise mechanisms by which they influence kidney function remained largely unexplored. Here, we examine the role of 1 GWAS-identified gene by creating mice deficient for Galnt11, which encodes a member of the enzyme family that initiates protein O-glycosylation, an essential posttranslational modification known to influence protein function and stability. We find that Galnt11-deficient mice display low-molecular-weight proteinuria and have specific defects in proximal tubule-mediated resorption of vitamin D binding protein, α1-microglobulin, and retinol binding protein. Moreover, we identify the endocytic receptor megalin (LRP2) as a direct target of Galnt11 in vivo. Megalin in Galnt11-deficient mice displays reduced ligand binding and undergoes age-related loss within the kidney. Differential mass spectrometry revealed specific sites of Galnt11-mediated glycosylation within mouse kidney megalin/LRP2 that are known to be involved in ligand binding, suggesting that O-glycosylation directly influences the ability to bind ligands. In support of this, recombinant megalin containing these sites displayed reduced albumin binding in cells deficient for Galnt11 Our results provide insight into the association between GALNT11 and CKD, and identify a role for Galnt11 in proper kidney function.


Subject(s)
Kidney/physiopathology , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Renal Insufficiency, Chronic/metabolism , Alpha-Globulins/genetics , Alpha-Globulins/metabolism , Animals , Endocytosis , Female , Glycosylation , Humans , Kidney/metabolism , Kidney Tubules, Proximal/metabolism , Ligands , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Male , Mice , Mice, Knockout , N-Acetylgalactosaminyltransferases/genetics , Protein Binding , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/physiopathology , Vitamin D-Binding Protein/genetics , Vitamin D-Binding Protein/metabolism
8.
J Biol Chem ; 295(5): 1411-1425, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31882545

ABSTRACT

The importance of the microbiome in health and its disruption in disease is continuing to be elucidated. However, the multitude of host and environmental factors that influence the microbiome are still largely unknown. Here, we examined UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferase 3 (Galnt3)-deficient mice, which serve as a model for the disease hyperphosphatemic familial tumoral calcinosis (HFTC). In HFTC, loss of GALNT3 activity in the bone is thought to lead to altered glycosylation of the phosphate-regulating hormone fibroblast growth factor 23 (FGF23), resulting in hyperphosphatemia and subdermal calcified tumors. However, GALNT3 is expressed in other tissues in addition to bone, suggesting that systemic loss could result in other pathologies. Using semiquantitative real-time PCR, we found that Galnt3 is the major O-glycosyltransferase expressed in the secretory cells of salivary glands. Additionally, 16S rRNA gene sequencing revealed that the loss of Galnt3 resulted in changes in the structure, composition, and stability of the oral microbiome. Moreover, we identified the major secreted salivary mucin, Muc10, as an in vivo substrate of Galnt3. Given that mucins and their O-glycans are known to interact with various microbes, our results suggest that loss of Galnt3 decreases glycosylation of Muc10, which alters the composition and stability of the oral microbiome. Considering that oral findings have been documented in HFTC patients, our study suggests that investigating GALNT3-mediated changes in the oral microbiome may be warranted.


Subject(s)
Calcinosis/metabolism , Calcinosis/microbiology , Hyperostosis, Cortical, Congenital/metabolism , Hyperostosis, Cortical, Congenital/microbiology , Hyperphosphatemia/metabolism , Hyperphosphatemia/microbiology , Microbiota/genetics , N-Acetylgalactosaminyltransferases/metabolism , Salivary Glands/metabolism , Animals , Calcinosis/genetics , Female , Fibroblast Growth Factor-23 , Glycosylation , Glycosyltransferases/metabolism , Hyperostosis, Cortical, Congenital/genetics , Hyperphosphatemia/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucins/chemistry , Mucins/metabolism , N-Acetylgalactosaminyltransferases/genetics , Polysaccharides/metabolism , RNA, Ribosomal, 16S/genetics , Polypeptide N-acetylgalactosaminyltransferase
9.
J Biol Chem ; 295(35): 12525-12536, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32669364

ABSTRACT

Mucin-type O-glycosylation is an essential post-translational modification required for protein secretion, extracellular matrix formation, and organ growth. O-Glycosylation is initiated by a large family of enzymes (GALNTs in mammals and PGANTs in Drosophila) that catalyze the addition of GalNAc onto the hydroxyl groups of serines or threonines in protein substrates. These enzymes contain two functional domains: a catalytic domain and a C-terminal ricin-like lectin domain comprised of three potential GalNAc recognition repeats termed α, ß, and γ. The catalytic domain is responsible for binding donor and acceptor substrates and catalyzing transfer of GalNAc, whereas the lectin domain recognizes more distant extant GalNAc on previously glycosylated substrates. We previously demonstrated a novel role for the α repeat of lectin domain in influencing charged peptide preferences. Here, we further interrogate how the differentially spliced α repeat of the PGANT9A and PGANT9B O-glycosyltransferases confers distinct preferences for a variety of endogenous substrates. Through biochemical analyses and in silico modeling using preferred substrates, we find that a combination of charged residues within the α repeat and charged residues in the flexible gating loop of the catalytic domain distinctively influence the peptide substrate preferences of each splice variant. Moreover, PGANT9A and PGANT9B also display unique glycopeptide preferences. These data illustrate how changes within the noncatalytic lectin domain can alter the recognition of both peptide and glycopeptide substrates. Overall, our results elucidate a novel mechanism for modulating substrate preferences of O-glycosyltransferases via alternative splicing within specific subregions of functional domains.


Subject(s)
Computer Simulation , Drosophila Proteins/chemistry , Glycopeptides/chemistry , Glycosyltransferases/chemistry , Alternative Splicing , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Glycopeptides/genetics , Glycosylation , Glycosyltransferases/genetics , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Substrate Specificity
10.
Glycoconj J ; 38(2): 145-156, 2021 04.
Article in English | MEDLINE | ID: mdl-33068214

ABSTRACT

O-glycosylation is a highly diverse and complex form of protein post-translational modification. Mucin-type O-glycosylation is initiated by the transfer of N-acetyl-galactosamine (GalNAc) to the hydroxyl group of serine, threonine and tyrosine residues through catalysis by a family of glycosyltransferases, the UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferases (E.C. 2.4.1.41) that are conserved across metazoans. In the last decade, structural characterization of glycosylation has substantially advanced due to the development of analytical methods and advances in mass spectrometry. However, O-glycosite mapping remains challenging since mucin-type O-glycans are densely packed, often protecting proteins from cleavage by proteases. Adding to the complexity is the fact that a given glycosite can be modified by different glycans, resulting in an array of glycoforms rising from one glycosite. In this study, we investigated conditions of solid phase extraction (SPE) enrichment, protease digestion, and Electron-transfer/Higher Energy Collision Dissociation (EThcD) fragmentation to optimize identification of O-glycosites in densely glycosylated proteins. Our results revealed that anion-exchange stationary phase is sufficient for glycopeptide enrichment; however, the use of a hydrophobic-containing sorbent was detrimental to the binding of polar-hydrophilic glycopeptides. Different proteases can be employed for enhancing coverage of O-glycosites, while derivatization of negatively charged amino acids or sialic acids would enhance the identification of a short O-glycopeptides. Using a longer than normal electron transfer dissociation (ETD) reaction time, we obtained enhanced coverage of peptide bonds that facilitated the localization of O-glycosites. O-glycosite mapping strategy via proteases, cut-off filtration and solid-phase chemoenzymatic processing. Glycopeptides are enriched by SPE column, followed by release of N-glycans, collection of higher MW O-glycopeptides via MW cut-off filter, O-glycopeptide release via O-protease, and finally detected by LC-MS/MS using EThcD.


Subject(s)
Glycopeptides/analysis , Glycopeptides/chemistry , Solid Phase Extraction/methods , Tandem Mass Spectrometry/methods , Amino Acids/chemistry , Animals , Cattle , Chemical Fractionation , Chromatography, Liquid , Fetuins/analysis , Fetuins/chemistry , Fetuins/metabolism , Glycopeptides/metabolism , Glycosylation , Mucins/analysis , Mucins/chemistry , Mucins/metabolism , N-Acetylneuraminic Acid/chemistry , Peptide Hydrolases/chemistry , Submandibular Gland/chemistry
11.
J Biol Chem ; 294(34): 12579-12580, 2019 08 23.
Article in English | MEDLINE | ID: mdl-31444307

ABSTRACT

Natriuretic peptides (NPs) are hormones involved in maintaining heart health that undergo proteolytic cleavage to become activated. Previous work has shown that O-GalNAc glycans affect their processing and activation. Here, Goetze, Schjoldager, and colleagues now provide comprehensive characterization of O-glycosylation of NPs, revealing that all members of the NP family can be modified by O-GalNAc glycans. Intriguingly, the study discovers glycans in the receptor-binding region of the A-type natriuretic peptide (ANP), demonstrating that they affect both stability and activity of ANP. These results may inform future therapeutic approaches for heart failure using peptide glycoforms.


Subject(s)
Heart Failure/metabolism , Natriuretic Peptides/metabolism , Polysaccharides/metabolism , Animals , Glycosylation , Humans
12.
J Biol Chem ; 294(51): 19498-19510, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31690624

ABSTRACT

Regulated secretion is a conserved process occurring across diverse cells and tissues. Current models suggest that the conserved cargo receptor Tango1 mediates the packaging of collagen into large coat protein complex II (COPII) vesicles that move from the endoplasmic reticulum (ER) to the Golgi apparatus. However, how Tango1 regulates the formation of COPII carriers and influences the secretion of other cargo remains unknown. Here, through high-resolution imaging of Tango1, COPII, Golgi, and secretory cargo (mucins) in Drosophila larval salivary glands, we found that Tango1 forms ring-like structures that mediate the formation of COPII rings rather than vesicles. These COPII rings act as docking sites for the cis-Golgi. Moreover, we observed nascent secretory mucins emerging from the Golgi side of these Tango1-COPII-Golgi complexes, suggesting that these structures represent functional docking sites/fusion points between the ER exit sites and the Golgi. Loss of Tango1 disrupted the formation of COPII rings, the association of COPII with the cis-Golgi, mucin O-glycosylation, and secretory granule biosynthesis. Additionally, we identified a Tango1 self-association domain that is essential for formation of this structure. Our results provide evidence that Tango1 organizes an interaction site where secretory cargo is efficiently transferred from the ER to Golgi and then to secretory vesicles. These findings may explain how the loss of Tango1 can influence Golgi/ER morphology and affect the secretion of diverse proteins across many tissues.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/embryology , Endoplasmic Reticulum/physiology , Gene Expression Regulation, Developmental , Golgi Apparatus/physiology , Secretory Vesicles/physiology , Animals , Animals, Genetically Modified , Binding Sites , COP-Coated Vesicles/metabolism , Glycosylation , Image Processing, Computer-Assisted , Protein Transport , RNA Interference , Salivary Glands/embryology
13.
J Biol Chem ; 293(4): 1315-1316, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29374084

ABSTRACT

Changes in the O-glycosylation of proteins have long been associated with the development of cancer, but establishing causal relationships between altered glycosylation and cancer progression remains incomplete. In this study, the authors perform comparative analyses of the cellular phenotypes, transcriptional changes, and alterations in the glycoproteome in colon cancer cells that differentially express one glycosyltransferase. Their results provide a wealth of data on which future studies can be based.


Subject(s)
Colonic Neoplasms/metabolism , Neoplasm Proteins/metabolism , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Glycosylation , Humans , Neoplasm Proteins/genetics
14.
J Cell Sci ; 130(8): 1355-1363, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28302911

ABSTRACT

Real-time imaging of regulated exocytosis in secreting organs can provide unprecedented temporal and spatial detail. Here, we highlight recent advances in 3D time-lapse imaging in Drosophila salivary glands at single-granule resolution. Using fluorescently labeled proteins expressed in the fly, it is now possible to image the dynamics of vesicle biogenesis and the cytoskeletal factors involved in secretion. 3D imaging over time allows one to visualize and define the temporal sequence of events, including clearance of cortical actin, fusion pore formation, mixing of the vesicular and plasma membranes and recruitment of components of the cytoskeleton. We will also discuss the genetic tools available in the fly that allow one to interrogate the essential factors involved in secretory vesicle formation, cargo secretion and the ultimate integration of the vesicular and plasma membranes. We argue that the combination of high-resolution real-time imaging and powerful genetics provides a platform to investigate the role of any factor in regulated secretion.


Subject(s)
Drosophila/physiology , Exocytosis , Salivary Glands/ultrastructure , Secretory Vesicles/ultrastructure , Time-Lapse Imaging/methods , Animals , Cytoskeleton/metabolism , Humans , Imaging, Three-Dimensional , Membrane Fusion , Microscopy, Fluorescence , Molecular Biology/methods , Salivary Glands/metabolism
15.
J Biol Chem ; 292(52): 21231-21242, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29127201

ABSTRACT

The mucous barrier of our digestive tract is the first line of defense against pathogens and damage. Disruptions in this barrier are associated with diseases such as Crohn's disease, colitis, and colon cancer, but mechanistic insights into these processes and diseases are limited. We have previously shown that loss of a conserved O-glycosyltransferase (PGANT4) in Drosophila results in aberrant secretion of components of the peritrophic/mucous membrane in the larval digestive tract. Here, we show that loss of PGANT4 disrupts the mucosal barrier, resulting in epithelial expression of the IL-6-like cytokine Upd3, leading to activation of JAK/STAT signaling, differentiation of cells that form the progenitor cell niche, and abnormal proliferation of progenitor cells. This niche disruption could be recapitulated by overexpressing upd3 and rescued by deleting upd3, highlighting a crucial role for this cytokine. Moreover, niche integrity and cell proliferation in pgant4-deficient animals could be rescued by overexpression of the conserved cargo receptor Tango1 and partially rescued by supplementation with exogenous mucins or treatment with antibiotics. Our findings help elucidate the paracrine signaling events activated by a compromised mucosal barrier and provide a novel in vivo screening platform for mucin mimetics and other strategies to treat diseases of the oral mucosa and digestive tract.


Subject(s)
Drosophila Proteins/metabolism , Intestinal Mucosa/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Stem Cell Niche/physiology , Animals , Cell Differentiation/physiology , Cytokines/metabolism , Drosophila/metabolism , Drosophila Proteins/genetics , Intestinal Mucosa/physiology , Janus Kinases/metabolism , N-Acetylgalactosaminyltransferases/genetics , Paracrine Communication/physiology , STAT Transcription Factors/metabolism , Signal Transduction/physiology , Stem Cells/cytology
16.
Proc Natl Acad Sci U S A ; 111(20): 7296-301, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24799692

ABSTRACT

Polarized secretion is crucial in many tissues. The conserved protein modification, O-glycosylation, plays a role in regulating secretion. However, the mechanisms by which this occurs are unknown. Here, we demonstrate that an O-glycosyltransferase functions as a novel regulator of secretion and secretory vesicle formation in vivo by glycosylating the essential Golgi/endoplasmic reticulum protein, Tango1 (Transport and Golgi organization 1), and conferring protection from furin-mediated proteolysis. Loss of the O-glycosyltransferase PGANT4 resulted in Tango1 cleavage, loss of secretory granules, and disrupted apical secretion. The secretory defects seen upon loss of pgant4 could be rescued either by overexpression of Tango1 or by knockdown of a specific furin (Dfur2) in vivo. Our studies elucidate a novel regulatory mechanism whereby secretion is influenced by the yin/yang of O-glycosylation and proteolytic cleavage. Moreover, our data have broader implications for the potential treatment of diseases resulting from the loss of O-glycosylation by modulating the activity of specific proteases.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Drosophila Proteins/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Subtilisins/metabolism , Animals , Calcinosis , Catalysis , Drosophila melanogaster , Endoplasmic Reticulum/metabolism , Glycosylation , Golgi Apparatus/metabolism , Mucins/metabolism , Mutation , Protein Binding , Protein Processing, Post-Translational , RNA Interference
17.
Glycobiology ; 26(4): 360-76, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26610890

ABSTRACT

A large family of UDP-GalNAc:polypeptide GalNAc transferases (ppGalNAc-Ts) initiates and defines sites of mucin-type Ser/Thr-O-GalNAc glycosylation. Family members have been classified into peptide- and glycopeptide-preferring subfamilies, although both families possess variable activities against glycopeptide substrates. All but one isoform contains a C-terminal carbohydrate-binding lectin domain whose roles in modulating glycopeptide specificity is just being understood. We have previously shown for several peptide-preferring isoforms that the presence of a remote Thr-O-GalNAc, 6-17 residues from a Ser/Thr acceptor site, may enhance overall catalytic activity in an N- or C-terminal direction. This enhancement varies with isoform and is attributed to Thr-O-GalNAc interactions at the lectin domain. We now report on the glycopeptide substrate utilization of a series of glycopeptide (human-ppGalNAc-T4, T7, T10, T12 and fly PGANT7) and peptide-preferring transferases (T2, T3 and T5) by exploiting a series of random glycopeptide substrates designed to probe the functions of their catalytic and lectin domains. Glycosylation was observed at the -3, -1 and +1 residues relative to a neighboring Thr-O-GalNAc, depending on isoform, which we attribute to specific Thr-O-GalNAc binding at the catalytic domain. Additionally, these glycopeptide-preferring isoforms show remote lectin domain-assisted Thr-O-GalNAc enhancements that vary from modest to none. We conclude that the glycopeptide specificity of the glycopeptide-preferring isoforms predominantly resides in their catalytic domain but may be further modulated by remote lectin domain interactions. These studies further demonstrate that both domains of the ppGalNAc-Ts have specialized and unique functions that work in concert to control and order mucin-type O-glycosylation.


Subject(s)
Glycopeptides/chemistry , Lectins/chemistry , Mucins/chemistry , Sialyltransferases/chemistry , Amino Acid Sequence/genetics , Binding Sites , Carbohydrates/chemistry , Carbohydrates/genetics , Catalytic Domain , Fucose/analogs & derivatives , Fucose/chemistry , Glycopeptides/biosynthesis , Glycopeptides/genetics , Glycosylation , Humans , Lectins/genetics , Mucins/biosynthesis , Mucins/genetics , Phylogeny , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sialyltransferases/genetics , Substrate Specificity
18.
J Biol Chem ; 288(10): 6921-9, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23329828

ABSTRACT

Mucin-type O-glycosylation is an evolutionarily conserved protein modification present on membrane-bound and secreted proteins. Aberrations in O-glycosylation are responsible for certain human diseases and are associated with disease risk factors. Recent studies have demonstrated essential roles for mucin-type O-glycosylation in protein secretion, stability, processing, and function. Here, we summarize our current understanding of the diverse roles of mucin-type O-glycosylation during eukaryotic development. Appreciating how this conserved modification operates in developmental processes will provide insight into its roles in human disease and disease susceptibilities.


Subject(s)
Glycosyltransferases/metabolism , Membrane Glycoproteins/metabolism , Mucins/metabolism , Polysaccharides/metabolism , Acetylgalactosamine/metabolism , Animals , Glycosylation , Humans , Models, Biological , N-Acetylgalactosaminyltransferases/metabolism
19.
J Biol Chem ; 287(8): 5243-52, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22157008

ABSTRACT

Mucin-type O-glycosylation represents a major form of post-translational modification that is conserved across most eukaryotic species. This type of glycosylation is initiated by a family of enzymes (GalNAc-Ts in mammals and PGANTs in Drosophila) whose members are expressed in distinct spatial and temporal patterns during development. Previous work from our group demonstrated that one member of this family is essential for viability and another member modulates extracellular matrix composition and integrin-mediated cell adhesion during development. To investigate whether other members of this family are essential, we employed RNA interference (RNAi) to each gene in vivo. Using this approach, we identified 4 additional pgant genes that are required for viability. Ubiquitous RNAi to pgant4, pgant5, pgant7, or the putative glycosyltransferase CG30463 resulted in lethality. Tissue-specific RNAi was also used to define the specific organ systems and tissues in which each essential family member is required. Interestingly, each essential pgant had a unique complement of tissues in which it was required. Additionally, certain tissues (mesoderm, digestive system, and tracheal system) required more than one pgant, suggesting unique functions for specific enzymes in these tissues. Expanding upon our RNAi results, we found that conventional mutations in pgant5 resulted in lethality and specific defects in specialized cells of the digestive tract, resulting in loss of proper digestive system acidification. In summary, our results highlight essential roles for O-glycosylation and specific members of the pgant family in many aspects of development and organogenesis.


Subject(s)
Drosophila melanogaster/enzymology , N-Acetylgalactosaminyltransferases/metabolism , Animals , Cell Adhesion , Digestive System/enzymology , Digestive System/growth & development , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Female , Glycosylation , Hydrogen-Ion Concentration , Male , Mutation , N-Acetylgalactosaminyltransferases/deficiency , N-Acetylgalactosaminyltransferases/genetics , Organ Specificity , Oxygen/metabolism , RNA Interference , Wings, Animal/cytology
20.
J Biol Chem ; 287(25): 20967-74, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22549772

ABSTRACT

O-Linked glycosylation is a functionally and structurally diverse type of protein modification present in many tissues and across many species. α-Dystroglycan (α-DG), a protein linked to the extracellular matrix, whose glycosylation status is associated with human muscular dystrophies, displays two predominant types of O-glycosylation, O-linked mannose (O-Man) and O-linked N-acetylgalactosamine (O-GalNAc), in its highly conserved mucin-like domain. The O-Man is installed by an enzyme complex present in the endoplasmic reticulum. O-GalNAc modifications are initiated subsequently in the Golgi apparatus by the UDP-GalNAc polypeptide N-acetylgalactosaminyltransferase (ppGalNAc-T) enzymes. How the presence and position of O-Man influences the action of the ppGalNAc-Ts on α-DG and the distribution of the two forms of glycosylation in this domain is not known. Here, we investigated the interplay between O-Man and the addition of O-GalNAc by examining the activity of the ppGalNAc-Ts on peptides and O-Man-containing glycopeptides mimicking those found in native α-DG. These synthetic glycopeptides emulate intermediate structures, not otherwise readily available from natural sources. Through enzymatic and mass spectrometric methods, we demonstrate that the presence and specific location of O-Man can impact either the regional exclusion or the site of O-GalNAc addition on α-DG, elucidating the factors contributing to the glycosylation patterns observed in vivo. These results provide evidence that one form of glycosylation can influence another form of glycosylation in α-DG and suggest that in the absence of proper O-mannosylation, as is associated with certain forms of muscular dystrophy, aberrant O-GalNAc modifications may occur and could play a role in disease presentation.


Subject(s)
Acetylgalactosamine/metabolism , Dystroglycans/metabolism , Mannose/metabolism , Multienzyme Complexes/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Uridine Diphosphate N-Acetylgalactosamine/metabolism , Acetylgalactosamine/genetics , Animals , Cell Line , Dystroglycans/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Glycosylation , Humans , Mannose/genetics , Mice , Multienzyme Complexes/genetics , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , N-Acetylgalactosaminyltransferases/genetics , Uridine Diphosphate N-Acetylgalactosamine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL