Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(43): e2303703120, 2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-37862385

RESUMEN

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.


Asunto(s)
Glicoproteínas , Enfermedades Metabólicas , Animales , Ratones , Glicosilación , Glicoproteínas/genética , Glicoproteínas/metabolismo , Proteoma/metabolismo , Polisacáridos , Polipéptido N-Acetilgalactosaminiltransferasa
3.
JAMA ; 329(20): 1740-1741, 2023 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-37115557

RESUMEN

This Viewpoint discusses specific areas of improvement in the National Institutes of Health's funding of and research criteria for clinical trials to be inclusive, transparent, and broad reaching.

5.
J Am Dent Assoc ; 154(1): 1-2, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36428128
7.
Mol Metab ; 60: 101472, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35304331

RESUMEN

OBJECTIVE: GALNT2, encoding polypeptide N-acetylgalactosaminyltransferase 2 (GalNAc-T2), was initially discovered as a regulator of high-density lipoprotein metabolism. GalNAc-T2 is known to exert these effects through post-translational modification, i.e., O-linked glycosylation of secreted proteins with established roles in plasma lipid metabolism. It has recently become clear that loss of GALNT2 in rodents, cattle, nonhuman primates, and humans should be regarded as a novel congenital disorder of glycosylation that affects development and body weight. The role of GALNT2 in metabolic abnormalities other than plasma lipids, including insulin sensitivity and energy homeostasis, is poorly understood. METHODS: GWAS data from the UK Biobank was used to study variation in the GALNT2 locus beyond changes in high-density lipoprotein metabolism. Experimental data were obtained through studies in Galnt2-/- mice and wild-type littermates on both control and high-fat diet. RESULTS: First, we uncovered associations between GALNT2 gene variation, adiposity, and body mass index in humans. In mice, we identify the insulin receptor as a novel substrate of GalNAc-T2 and demonstrate that Galnt2-/- mice exhibit decreased adiposity, alterations in insulin signaling and a shift in energy substrate utilization in the inactive phase. CONCLUSIONS: This study identifies a novel role for GALNT2 in energy homeostasis, and our findings suggest that the local effects of GalNAc-T2 are mediated through posttranslational modification of the insulin receptor.


Asunto(s)
Lipoproteínas HDL , Receptor de Insulina , Animales , Bovinos , Glicosilación , Homeostasis , Ratones , N-Acetilgalactosaminiltransferasas , Polipéptido N-Acetilgalactosaminiltransferasa
9.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34732583

RESUMEN

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.


Asunto(s)
SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Animales , Fusión Celular , Línea Celular , Furina/metabolismo , Células Gigantes , Glicosilación , Humanos , N-Acetilgalactosaminiltransferasas/metabolismo , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Polipéptido N-Acetilgalactosaminiltransferasa
10.
Science ; 373(6551): 165-167, 2021 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-34244402
11.
Cell ; 184(12): 3075-3079, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34115967

RESUMEN

NIH has acknowledged and committed to ending structural racism. The framework for NIH's approach, summarized here, includes understanding barriers; developing robust health disparities/equity research; improving its internal culture; being transparent and accountable; and changing the extramural ecosystem so that diversity, equity, and inclusion are reflected in funded research and the biomedical workforce.


Asunto(s)
Investigación Biomédica , National Institutes of Health (U.S.) , Racismo Sistemático , Diversidad Cultural , Humanos , Apoyo a la Investigación como Asunto/economía , Estados Unidos
12.
bioRxiv ; 2021 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-33564758

RESUMEN

The SARS-CoV-2 coronavirus responsible for the global pandemic contains a unique furin cleavage site in the spike protein (S) that increases viral infectivity and syncytia formation. Here, we show that O-glycosylation near the furin cleavage site is mediated by specific members of the GALNT enzyme family and is dependent on the novel proline at position 681 (P681). We further demonstrate that O-glycosylation of S decreases furin cleavage. 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 O-glycosylation may influence viral infectivity/tropism by modulating furin cleavage of S and provide mechanistic insight into the potential role of P681 mutations in the recently identified, highly transmissible B.1.1.7 variant.

13.
Glycoconj J ; 38(2): 145-156, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33068214

RESUMEN

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.


Asunto(s)
Glicopéptidos/análisis , Glicopéptidos/química , Extracción en Fase Sólida/métodos , Espectrometría de Masas en Tándem/métodos , Aminoácidos/química , Animales , Bovinos , Fraccionamiento Químico , Cromatografía Liquida , Fetuínas/análisis , Fetuínas/química , Fetuínas/metabolismo , Glicopéptidos/metabolismo , Glicosilación , Mucinas/análisis , Mucinas/química , Mucinas/metabolismo , Ácido N-Acetilneuramínico/química , Péptido Hidrolasas/química , Glándula Submandibular/química
14.
J Biol Chem ; 295(35): 12525-12536, 2020 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-32669364

RESUMEN

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.


Asunto(s)
Simulación por Computador , Proteínas de Drosophila/química , Glicopéptidos/química , Glicosiltransferasas/química , Empalme Alternativo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster , Glicopéptidos/genética , Glicosilación , Glicosiltransferasas/genética , Humanos , Isoenzimas/química , Isoenzimas/genética , Especificidad por Sustrato
15.
Proc Natl Acad Sci U S A ; 116(50): 25196-25202, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31740596

RESUMEN

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.


Asunto(s)
Riñón/fisiopatología , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , N-Acetilgalactosaminiltransferasas/metabolismo , Insuficiencia Renal Crónica/metabolismo , alfa-Globulinas/genética , alfa-Globulinas/metabolismo , Animales , Endocitosis , Femenino , Glicosilación , Humanos , Riñón/metabolismo , Túbulos Renales Proximales/metabolismo , Ligandos , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Masculino , Ratones , Ratones Noqueados , N-Acetilgalactosaminiltransferasas/genética , Unión Proteica , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/fisiopatología , Proteína de Unión a Vitamina D/genética , Proteína de Unión a Vitamina D/metabolismo
16.
Proc Natl Acad Sci U S A ; 116(41): 20404-20410, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31548401

RESUMEN

Polypeptide N-acetylgalactosaminyl transferases (GalNAc-Ts) initiate mucin type O-glycosylation by catalyzing the transfer of N-acetylgalactosamine (GalNAc) to Ser or Thr on a protein substrate. Inactive and partially active variants of the isoenzyme GalNAc-T12 are present in subsets of patients with colorectal cancer, and several of these variants alter nonconserved residues with unknown functions. While previous biochemical studies have demonstrated that GalNAc-T12 selects for peptide and glycopeptide substrates through unique interactions with its catalytic and lectin domains, the molecular basis for this distinct substrate selectivity remains elusive. Here we examine the molecular basis of the activity and substrate selectivity of GalNAc-T12. The X-ray crystal structure of GalNAc-T12 in complex with a di-glycosylated peptide substrate reveals how a nonconserved GalNAc binding pocket in the GalNAc-T12 catalytic domain dictates its unique substrate selectivity. In addition, the structure provides insight into how colorectal cancer mutations disrupt the activity of GalNAc-T12 and illustrates how the rules dictating GalNAc-T12 function are distinct from those for other GalNAc-Ts.


Asunto(s)
Neoplasias Colorrectales/metabolismo , N-Acetilgalactosaminiltransferasas/química , N-Acetilgalactosaminiltransferasas/metabolismo , Proteínas de Neoplasias/química , Secuencia de Aminoácidos , Humanos , Modelos Moleculares , Conformación Proteica
17.
Science ; 363(6433): 1292-1294, 2019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30898925
18.
Cancer J ; 24(3): 111-114, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30273184

RESUMEN

As a part of the Cancer Moonshot, the National Cancer Institute, part of the National Institutes of Health, the Foundation for National Institutes of Health, the US Food and Drug Administration, and 12 pharmaceutical companies have formed a 5-year, $220 million precompetitive public-private research collaboration called the Partnership for Accelerating Cancer Therapies. A systematic cross-sector effort to identify and develop robust, standardized biomarkers and related clinical data, Partnership for Accelerating Cancer Therapies will support the selection and testing of promising immunotherapies for the treatment of cancer, with the goal of bringing effective therapy to more patients.


Asunto(s)
Neoplasias/economía , Neoplasias/terapia , Biomarcadores de Tumor/metabolismo , Humanos , National Cancer Institute (U.S.)/economía , Neoplasias/metabolismo , Estados Unidos , United States Food and Drug Administration/economía
19.
Nat Commun ; 9(1): 3508, 2018 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-30158631

RESUMEN

Regulated secretion is an essential process where molecules destined for export are directed to membranous secretory granules, where they undergo packaging and maturation. Here, we identify a gene (pgant9) that influences the structure and shape of secretory granules within the Drosophila salivary gland. Loss of pgant9, which encodes an O-glycosyltransferase, results in secretory granules with an irregular, shard-like morphology, and altered glycosylation of cargo. Interestingly, pgant9 undergoes a splicing event that acts as a molecular switch to alter the charge of a loop controlling access to the active site of the enzyme. The splice variant with the negatively charged loop glycosylates the positively charged secretory cargo and rescues secretory granule morphology. Our study highlights a mechanism for dictating substrate specificity within the O-glycosyltransferase enzyme family. Moreover, our in vitro and in vivo studies suggest that the glycosylation status of secretory cargo influences the morphology of maturing secretory granules.


Asunto(s)
Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Vesículas Secretoras/metabolismo , Animales , Drosophila , Proteínas de Drosophila/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Vesículas Secretoras/genética , Especificidad por Sustrato
20.
Glycobiology ; 28(11): 841-848, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30084948

RESUMEN

Mucin-type O-glycosylation is an evolutionarily conserved and essential post-translational protein modification that is initiated in the Golgi apparatus by a family of enzymes known as the UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferases (GalNAc-Ts). GalNAc-Ts are type II membrane proteins which contain short N-terminal tails located in the cytoplasm, a transmembrane domain that crosses the Golgi membrane, to which is connected a stem region that tethers the C-terminal catalytic and lectin domains that reside in the Golgi lumen. Although mucin-type O-glycans have been shown to play critical roles in numerous biological processes, little is known about how the GalNAc-Ts are targeted to their site of action within the Golgi complex. Here, we investigate the essential protein domains required for Golgi localization of four representative members of the GalNAc-T family of enzymes. We find that GalNAc-T1 and -T2 require their cytoplasmic tail and transmembrane domains for proper Golgi localization, while GalNAc-T10 requires its transmembrane and luminal stem domains. GalNAc-T7 can use either its cytoplasmic tail or its luminal stem, in combination with its transmembrane domain, to localize to the Golgi. We determined that a single glutamic acid in the GalNAc-T10 cytoplasmic tail inhibits its ability to localize to the Golgi via a cytoplasmic tail-dependent mechanism. We therefore demonstrate that despite their similarity, different members of this enzyme family are directed to the Golgi by more than one set of targeting signals.


Asunto(s)
Aparato de Golgi/metabolismo , N-Acetilgalactosaminiltransferasas/metabolismo , Células Cultivadas , Humanos , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...