Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Dis Model Mech ; 17(4)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38566589

RESUMEN

The addition of O-linked ß-N-acetylglucosamine (O-GlcNAc) to proteins (referred to as O-GlcNAcylation) is a modification that is crucial for vertebrate development. O-GlcNAcylation is catalyzed by O-GlcNAc transferase (OGT) and reversed by O-GlcNAcase (OGA). Missense variants of OGT have recently been shown to segregate with an X-linked syndromic form of intellectual disability, OGT-linked congenital disorder of glycosylation (OGT-CDG). Although the existence of OGT-CDG suggests that O-GlcNAcylation is crucial for neurodevelopment and/or cognitive function, the underlying pathophysiologic mechanisms remain unknown. Here we report a mouse line that carries a catalytically impaired OGT-CDG variant. These mice show altered O-GlcNAc homeostasis with decreased global O-GlcNAcylation and reduced levels of OGT and OGA in the brain. Phenotypic characterization of the mice revealed lower body weight associated with reduced body fat mass, short stature and microcephaly. This mouse model will serve as an important tool to study genotype-phenotype correlations in OGT-CDG in vivo and for the development of possible treatment avenues for this disorder.


Asunto(s)
Modelos Animales de Enfermedad , Discapacidad Intelectual , N-Acetilglucosaminiltransferasas , Animales , N-Acetilglucosaminiltransferasas/metabolismo , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/deficiencia , Discapacidad Intelectual/genética , Encéfalo/patología , Encéfalo/metabolismo , Fenotipo , Ratones , Trastornos del Neurodesarrollo/patología , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/enzimología , beta-N-Acetilhexosaminidasas/metabolismo , Glicosilación , Peso Corporal
2.
Biochem Biophys Res Commun ; 589: 123-130, 2022 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-34906902

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is the most common liver disorder with intricate etiology. It is closely associated with metabolic syndrome, insulin resistance and endoplasmic reticulum (ER) stress. Exostosin1 (Ext1) is an ER-resident transmembrane glycosyltransferase, which plays an important role in ER homeostasis. Loss-of-function mutations in Ext1 link to hereditary multiple exostosis (HME). The present research was undertaken to identify the effect of Ext1 in the progress of NAFLD. High-fat-diet induced mice obesity, hepatic steatosis and decreased hepatic Ext1 expression. In consistent with evaluation of NAFLD mice possessing down-regulated Ext1 expression, free fatty acid (FFA) treatment blunted Ext1 expression in hepatocytes. In human subjects, HME patients presented elevated fasting blood glucose-one of the criteria that define insulin resistance. In vitro experiments, Ext1 deficiency promoted FFA-induced insulin resistance in hepatocytes by analysis of glycogen storage and hallmarks of gluconeogenesis, ascertaining its association with insulin resistance. Mechanically, Ext1 silencing exacerbated ER stress triggered by FFA, which severely disrupted autophagy in hepatocytes, and thereby accelerated the progression of NAFLD. In conclusion, our study demonstrates a beneficial role for Ext1 during the development of NAFLD, which establishes a novel correlation between Ext1 and ER stress-induced perturbations of autophagy during NAFLD progression.


Asunto(s)
N-Acetilglucosaminiltransferasas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/etiología , Obesidad/complicaciones , Animales , Autofagia , Línea Celular , Regulación hacia Abajo , Estrés del Retículo Endoplásmico , Ácidos Grasos no Esterificados/metabolismo , Silenciador del Gen , Gluconeogénesis , Hepatocitos/enzimología , Hepatocitos/patología , Resistencia a la Insulina , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , N-Acetilglucosaminiltransferasas/deficiencia
3.
Int J Mol Sci ; 22(16)2021 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-34445285

RESUMEN

N-glycosylation is essential for many biological processes in mammals. A variety of N-glycan structures exist, of which, the formation of bisecting N-acetylglucosamine (GlcNAc) is catalyzed by N-acetylglucosaminyltransferase-III (GnT-III, encoded by the Mgat3 gene). We previously identified various bisecting GlcNAc-modified proteins involved in Alzheimer's disease and cancer. However, the mechanisms by which GnT-III acts on the target proteins are unknown. Here, we performed comparative glycoproteomic analyses using brain membranes of wild type (WT) and Mgat3-deficient mice. Target glycoproteins of GnT-III were enriched with E4-phytohemagglutinin (PHA) lectin, which recognizes bisecting GlcNAc, and analyzed by liquid chromatograph-mass spectrometry. We identified 32 N-glycosylation sites (Asn-Xaa-Ser/Thr, Xaa ≠ Pro) that were modified with bisecting GlcNAc. Sequence alignment of identified N-glycosylation sites that displayed bisecting GlcNAc suggested that GnT-III does not recognize a specific primary amino acid sequence. The molecular modeling of GluA1 as one of the good cell surface substrates for GnT-III in the brain, indicated that GnT-III acts on N-glycosylation sites located in a highly flexible and mobile loop of GluA1. These results suggest that the action of GnT-III is partially affected by the tertiary structure of target proteins, which can accommodate bisecting GlcNAc that generates a bulky flipped-back conformation of the modified glycans.


Asunto(s)
Acetilglucosamina/metabolismo , Encéfalo/metabolismo , Membrana Celular/metabolismo , Péptidos/metabolismo , Receptores AMPA/metabolismo , Análisis de Secuencia de Proteína , Acetilglucosamina/genética , Animales , Membrana Celular/genética , Glicosilación , Ratones , Ratones Noqueados , N-Acetilglucosaminiltransferasas/deficiencia , N-Acetilglucosaminiltransferasas/metabolismo , Mapeo Peptídico , Péptidos/genética , Receptores AMPA/genética
4.
J Biol Chem ; 297(1): 100887, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34146542

RESUMEN

Liver fibrosis is a common characteristic of chronic liver diseases. The activation of hepatic stellate cells (HSCs) plays a key role in fibrogenesis in response to liver injury, yet the mechanism by which damaged hepatocytes modulate the activation of HSCs is poorly understood. Our previous studies have established that liver-specific deletion of O-GlcNAc transferase (OGT)leads to hepatocyte necroptosis and spontaneous fibrosis. Here, we report that OGT-deficient hepatocytes secrete trefoil factor 2 (TFF2) that activates HSCs and contributes to the fibrogenic process. The expression and secretion of TFF2 are induced in OGT-deficient hepatocytes but not in WT hepatocytes. TFF2 activates the platelet-derived growth factor receptor beta signaling pathway that promotes the proliferation and migration of primary HSCs. TFF2 protein expression is elevated in mice with carbon tetrachloride-induced liver injury. These findings identify TFF2 as a novel factor that mediates intercellular signaling between hepatocytes and HSCs and suggest a role of the hepatic OGT-TFF2 axis in the process of fibrogenesis.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Cirrosis Hepática/metabolismo , Factor Trefoil-2/metabolismo , Animales , Tetracloruro de Carbono/toxicidad , Línea Celular , Células Cultivadas , Exocitosis , Células Estrelladas Hepáticas/patología , Hepatocitos/patología , Humanos , Cirrosis Hepática/etiología , Ratones , N-Acetilglucosaminiltransferasas/deficiencia , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , Necroptosis , Transducción de Señal , Factor Trefoil-2/genética
5.
Am J Med Genet A ; 185(10): 3104-3110, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34089299

RESUMEN

Spondyloepimetaphyseal dysplasia (SEMD) is a group of genetic skeletal disorders characterized by disproportionate short stature, and varying degrees of vertebral, epiphyseal, and metaphyseal involvement of the skeleton. According to the Nosology and classification of genetic skeletal disorders 2019 revision, more than 20 types of SEMD have been identified, and SEMD with immune deficiency, EXTL3 type is one of the newcomers. Affected individuals display variable skeletal abnormalities and neurodevelopmental findings. Liver and kidney cysts have also been reported frequently. Patients may exhibit varying degrees of immune deficiency as well. To date, only 14 patients from 9 unrelated families with SEMD with immune deficiency, EXTL3 type have been reported in the literature. We report a new patient who is currently 15 years old in whom cystic liver lesions were detected in the prenatal period. Disproportionate short stature, mild developmental delay and a T- NK+ B+ immunological profile were detected in the postnatal follow-up. Exome sequence analysis revealed a previously reported homozygous missense variant in exon 3 c.953C > T; p.(Pro318Leu) in EXTL3.


Asunto(s)
Enfermedades Autoinmunes/genética , Predisposición Genética a la Enfermedad , Síndromes de Inmunodeficiencia/genética , N-Acetilglucosaminiltransferasas/genética , Osteocondrodisplasias/genética , Adolescente , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Niño , Preescolar , Enanismo/genética , Enanismo/patología , Femenino , Pruebas Genéticas , Homocigoto , Humanos , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/patología , Lactante , Masculino , Mutación Missense/genética , N-Acetilglucosaminiltransferasas/deficiencia , Osteocondrodisplasias/inmunología , Osteocondrodisplasias/patología , Linaje , Columna Vertebral/patología , Adulto Joven
6.
Cell Rep ; 34(13): 108905, 2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33789105

RESUMEN

Ogt catalyzed O-linked N-acetylglucosamine (O-GlcNAcylation, O-GlcNAc) plays an important function in diverse biological processes and diseases. However, the roles of Ogt in regulating neurogenesis remain largely unknown. Here, we show that Ogt deficiency or depletion in adult neural stem/progenitor cells (aNSPCs) leads to the diminishment of the aNSPC pool and aberrant neurogenesis and consequently impairs cognitive function in adult mice. RNA sequencing reveals that Ogt deficiency alters the transcription of genes relating to cell cycle, neurogenesis, and neuronal development. Mechanistic studies show that Ogt directly interacts with Notch1 and catalyzes the O-GlcNAc modification of Notch TM/ICD fragment. Decreased O-GlcNAc modification of TM/ICD increases the binding of E3 ubiquitin ligase Itch to TM/ICD and promotes its degradation. Itch knockdown rescues neurogenic defects induced by Ogt deficiency in vitro and in vivo. Our findings reveal the essential roles and mechanisms of Ogt and O-GlcNAc modification in regulating mammalian neurogenesis and cognition.


Asunto(s)
Envejecimiento/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Neurogénesis , Receptores Notch/metabolismo , Transducción de Señal , Células Madre/enzimología , Acetilglucosamina/metabolismo , Animales , Biocatálisis , Diferenciación Celular , Proliferación Celular , Eliminación de Gen , Glicosilación , Células HEK293 , Humanos , Memoria , Ratones Transgénicos , N-Acetilglucosaminiltransferasas/deficiencia , Proteolisis , Receptores Notch/química , Células Madre/citología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
7.
J Biol Chem ; 296: 100433, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33610554

RESUMEN

Defects in protein O-mannosylation lead to severe congenital muscular dystrophies collectively known as α-dystroglycanopathy. A hallmark of these diseases is the loss of the O-mannose-bound matriglycan on α-dystroglycan, which reduces cell adhesion to the extracellular matrix. Mutations in protein O-mannose ß1,2-N-acetylglucosaminyltransferase 1 (POMGNT1), which is crucial for the elongation of O-mannosyl glycans, have mainly been associated with muscle-eye-brain (MEB) disease. In addition to defects in cell-extracellular matrix adhesion, aberrant cell-cell adhesion has occasionally been observed in response to defects in POMGNT1. However, specific molecular consequences of POMGNT1 deficiency on cell-cell adhesion are largely unknown. We used POMGNT1 knockout HEK293T cells and fibroblasts from an MEB patient to gain deeper insight into the molecular changes in POMGNT1 deficiency. Biochemical and molecular biological techniques combined with proteomics, glycoproteomics, and glycomics revealed that a lack of POMGNT1 activity strengthens cell-cell adhesion. We demonstrate that the altered intrinsic adhesion properties are due to an increased abundance of N-cadherin (N-Cdh). In addition, site-specific changes in the N-glycan structures in the extracellular domain of N-Cdh were detected, which positively impact on homotypic interactions. Moreover, in POMGNT1-deficient cells, ERK1/2 and p38 signaling pathways are activated and transcriptional changes that are comparable with the epithelial-mesenchymal transition (EMT) are triggered, defining a possible molecular mechanism underlying the observed phenotype. Our study indicates that changes in cadherin-mediated cell-cell adhesion and other EMT-related processes may contribute to the complex clinical symptoms of MEB or α-dystroglycanopathy in general and suggests that the impact of changes in O-mannosylation on N-glycosylation has been underestimated.


Asunto(s)
Adhesión Celular/fisiología , N-Acetilglucosaminiltransferasas/deficiencia , N-Acetilglucosaminiltransferasas/metabolismo , Antígenos CD/metabolismo , Antígenos CD/fisiología , Cadherinas/metabolismo , Cadherinas/fisiología , Adhesión Celular/genética , Distroglicanos/metabolismo , Glicómica , Glicosilación , Glicosiltransferasas/deficiencia , Glicosiltransferasas/metabolismo , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Manosa/química , Distrofias Musculares/genética , N-Acetilglucosaminiltransferasas/fisiología , Polisacáridos , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33419956

RESUMEN

O-GlcNAc transferase (OGT), found in the nucleus and cytoplasm of all mammalian cell types, is essential for cell proliferation. Why OGT is required for cell growth is not known. OGT performs two enzymatic reactions in the same active site. In one, it glycosylates thousands of different proteins, and in the other, it proteolytically cleaves another essential protein involved in gene expression. Deconvoluting OGT's myriad cellular roles has been challenging because genetic deletion is lethal; complementation methods have not been established. Here, we developed approaches to replace endogenous OGT with separation-of-function variants to investigate the importance of OGT's enzymatic activities for cell viability. Using genetic complementation, we found that OGT's glycosyltransferase function is required for cell growth but its protease function is dispensable. We next used complementation to construct a cell line with degron-tagged wild-type OGT. When OGT was degraded to very low levels, cells stopped proliferating but remained viable. Adding back catalytically inactive OGT rescued growth. Therefore, OGT has an essential noncatalytic role that is necessary for cell proliferation. By developing a method to quantify how OGT's catalytic and noncatalytic activities affect protein abundance, we found that OGT's noncatalytic functions often affect different proteins from its catalytic functions. Proteins involved in oxidative phosphorylation and the actin cytoskeleton were especially impacted by the noncatalytic functions. We conclude that OGT integrates both catalytic and noncatalytic functions to control cell physiology.


Asunto(s)
Proliferación Celular/genética , Fibroblastos/metabolismo , Factor C1 de la Célula Huésped/genética , N-Acetilglucosaminiltransferasas/genética , Animales , Fibroblastos/citología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Ontología de Genes , Prueba de Complementación Genética , Glicosilación , Células HEK293 , Factor C1 de la Célula Huésped/metabolismo , Humanos , Redes y Vías Metabólicas/genética , Ratones , Anotación de Secuencia Molecular , N-Acetilglucosaminiltransferasas/deficiencia , Proteolisis
9.
J Biol Chem ; 296: 100297, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33460647

RESUMEN

The nutrient sensor O-GlcNAc transferase (OGT) catalyzes posttranslational addition of O-GlcNAc onto target proteins, influencing signaling pathways in response to cellular nutrient levels. OGT is highly expressed in pancreatic glucagon-secreting cells (α-cells), which secrete glucagon in response to hypoglycemia. The objective of this study was to determine whether OGT is necessary for the regulation of α-cell mass and function in vivo. We utilized genetic manipulation to produce two α-cell specific OGT-knockout models: a constitutive glucagon-Cre (αOGTKO) and an inducible glucagon-Cre (i-αOGTKO), which effectively delete OGT in α-cells. Using approaches including immunoblotting, immunofluorescent imaging, and metabolic phenotyping in vivo, we provide the first insight on the role of O-GlcNAcylation in α-cell mass and function. αOGTKO mice demonstrated normal glucose tolerance and insulin sensitivity but displayed significantly lower glucagon levels during both fed and fasted states. αOGTKO mice exhibited significantly lower α-cell glucagon content and α-cell mass at 6 months of age. In fasting, αOGTKO mice showed impaired pyruvate stimulated gluconeogenesis in vivo and reduced glucagon secretion in vitro. i-αOGTKO mice showed similarly reduced blood glucagon levels, defective in vitro glucagon secretion, and normal α-cell mass. Interestingly, both αOGTKO and i-αOGTKO mice had no deficiency in maintaining blood glucose homeostasis under fed or fasting conditions, despite impairment in α-cell mass and function, and glucagon content. In conclusion, these studies provide a first look at the role of OGT signaling in the α-cell, its effect on α-cell mass, and its importance in regulating glucagon secretion in hypoglycemic conditions.


Asunto(s)
Glucemia/metabolismo , Células Secretoras de Glucagón/enzimología , Glucagón/biosíntesis , N-Acetilglucosaminiltransferasas/genética , Obesidad/genética , Acilación/efectos de los fármacos , Animales , Ayuno/metabolismo , Femenino , Efecto Fundador , Glucagón/deficiencia , Células Secretoras de Glucagón/efectos de los fármacos , Células Secretoras de Glucagón/patología , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/genética , Prueba de Tolerancia a la Glucosa , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Resistencia a la Insulina , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Noqueados , N-Acetilglucosaminiltransferasas/deficiencia , Obesidad/enzimología , Obesidad/patología , Ácido Pirúvico/metabolismo , Ácido Pirúvico/farmacología
10.
PLoS One ; 15(11): e0242250, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33253217

RESUMEN

RATIONALE: The beta-O-linkage of N-acetylglucosamine (i.e., O-GlcNAc) to proteins is a pro-adaptive response to cellular insults. To this end, increased protein O-GlcNAcylation improves short-term survival of cardiomyocytes subjected to acute injury. This observation has been repeated by multiple groups and in multiple models; however, whether increased protein O-GlcNAcylation plays a beneficial role in more chronic settings remains an open question. OBJECTIVE: Here, we queried whether increasing levels of cardiac protein O-GlcNAcylation would be beneficial during infarct-induced heart failure. METHODS AND RESULTS: To achieve increased protein O-GlcNAcylation, we targeted Oga, the gene responsible for removing O-GlcNAc from proteins. Here, we generated mice with cardiomyocyte-restricted, tamoxifen-inducible haploinsufficient Oga gene. In the absence of infarction, we observed a slight reduction in ejection fraction in Oga deficient mice. Overall, Oga reduction had no major impact on ventricular function. In additional cohorts, mice of both sexes and both genotypes were subjected to infarct-induced heart failure and followed for up to four weeks, during which time cardiac function was assessed via echocardiography. Contrary to our prediction, the Oga deficient mice exhibited exacerbated-not improved-cardiac function at one week following infarction. When the observation was extended to 4 wk post-MI, this acute exacerbation was lost. CONCLUSIONS: The present findings, coupled with our previous work, suggest that altering the ability of cardiomyocytes to either add or remove O-GlcNAc modifications to proteins exacerbates early infarct-induced heart failure. We speculate that more nuanced approaches to regulating O-GlcNAcylation are needed to understand its role-and, in particular, the possibility of cycling, in the pathophysiology of the failing heart.


Asunto(s)
Infarto del Miocardio/patología , Miocardio/enzimología , N-Acetilglucosaminiltransferasas/genética , Disfunción Ventricular/etiología , Animales , Ecocardiografía , Femenino , Glicosilación , Haploinsuficiencia , Corazón/fisiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Infarto del Miocardio/complicaciones , Miocardio/metabolismo , Miocardio/patología , N-Acetilglucosaminiltransferasas/deficiencia , N-Acetilglucosaminiltransferasas/metabolismo , Tamoxifeno/farmacología , Regulación hacia Arriba , Función Ventricular/efectos de los fármacos
11.
Oxid Med Cell Longev ; 2020: 5602396, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32850000

RESUMEN

Since both O-GlcNAcylation and autophagy sense intracellular nutrient level, the alteration of those two pathways plays substantial roles in the progression of heart failure. Hence, determining the relationship between O-GlcNAcylation and autophagy is imperative to understand, prevent, and treat heart failure. However, the mechanism on how O-GlcNAcylation regulates autophagy in the heart is poorly investigated. In this study, we demonstrated that O-GlcNAcylation is required for autophagy in cardiomyocytes by utilizing an O-linked ß-N-acetylglucosamine transferase (OGT) cardiomyocyte-specific knockout mouse model for the first time. We also identified that OGT might regulate the initiation of autophagy in cardiomyocytes through promoting the activity of ULK1 by O-GlcNAcylation. In conclusion, our findings provide new insights into the molecular mechanisms underlying heart dysfunction and benefit the development of treatments for heart failure.


Asunto(s)
Autofagia , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Eliminación de Gen , Glicosilación , Ratones Endogámicos C57BL , Miocitos Cardíacos/enzimología , N-Acetilglucosaminiltransferasas/deficiencia , N-Acetilglucosaminiltransferasas/metabolismo
12.
J Inherit Metab Dis ; 43(6): 1333-1348, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32681751

RESUMEN

Asparagine-linked glycosylation 13 homolog (ALG13) encodes a nonredundant, highly conserved, X-linked uridine diphosphate (UDP)-N-acetylglucosaminyltransferase required for the synthesis of lipid linked oligosaccharide precursor and proper N-linked glycosylation. De novo variants in ALG13 underlie a form of early infantile epileptic encephalopathy known as EIEE36, but given its essential role in glycosylation, it is also considered a congenital disorder of glycosylation (CDG), ALG13-CDG. Twenty-four previously reported ALG13-CDG cases had de novo variants, but surprisingly, unlike most forms of CDG, ALG13-CDG did not show the anticipated glycosylation defects, typically detected by altered transferrin glycosylation. Structural homology modeling of two recurrent de novo variants, p.A81T and p.N107S, suggests both are likely to impact the function of ALG13. Using a corresponding ALG13-deficient yeast strain, we show that expressing yeast ALG13 with either of the highly conserved hotspot variants rescues the observed growth defect, but not its glycosylation abnormality. We present molecular and clinical data on 29 previously unreported individuals with de novo variants in ALG13. This more than doubles the number of known cases. A key finding is that a vast majority of the individuals presents with West syndrome, a feature shared with other CDG types. Among these, the initial epileptic spasms best responded to adrenocorticotropic hormone or prednisolone, while clobazam and felbamate showed promise for continued epilepsy treatment. A ketogenic diet seems to play an important role in the treatment of these individuals.


Asunto(s)
Trastornos Congénitos de Glicosilación/genética , N-Acetilglucosaminiltransferasas/deficiencia , N-Acetilglucosaminiltransferasas/genética , Espasmos Infantiles/genética , Biomarcadores , Preescolar , Trastornos Congénitos de Glicosilación/diagnóstico , Dieta Cetogénica , Femenino , Glicosilación , Humanos , Lactante , Masculino , Mutación , N-Acetilglucosaminiltransferasas/química , Espasmos Infantiles/diagnóstico , Transferrina/metabolismo
13.
J Biol Chem ; 295(25): 8560-8574, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32376684

RESUMEN

Epidermal growth factor (EGF) domain-specific O-GlcNAc transferase (EOGT) is an endoplasmic reticulum (ER)-resident protein that modifies EGF repeats of Notch receptors and thereby regulates Delta-like ligand-mediated Notch signaling. Several EOGT mutations that may affect putative N-glycosylation consensus sites are recorded in the cancer database, but the presence and function of N-glycans in EOGT have not yet been characterized. Here, we identified N-glycosylation sites in mouse EOGT and elucidated their molecular functions. Three predicted N-glycosylation consensus sequences on EOGT are highly conserved among mammalian species. Within these sites, we found that Asn-263 and Asn-354, but not Asn-493, are modified with N-glycans. Lectin blotting, endoglycosidase H digestion, and MS analysis revealed that both residues are modified with oligomannose N-glycans. Loss of an individual N-glycan on EOGT did not affect its endoplasmic reticulum (ER) localization, enzyme activity, and ability to O-GlcNAcylate Notch1 in HEK293T cells. However, simultaneous substitution of both N-glycosylation sites affected both EOGT maturation and expression levels without an apparent change in enzymatic activity, suggesting that N-glycosylation at a single site is sufficient for EOGT maturation and expression. Accordingly, a decrease in O-GlcNAc stoichiometry was observed in Notch1 co-expressed with an N263Q/N354Q variant compared with WT EOGT. Moreover, the N263Q/N354Q variant exhibited altered subcellular distribution within the ER in HEK293T cells, indicating that N-glycosylation of EOGT is required for its ER localization at the cell periphery. These results suggest critical roles of N-glycans in sustaining O-GlcNAc transferase function both by maintaining EOGT levels and by ensuring its proper subcellular localization in the ER.


Asunto(s)
Retículo Endoplásmico/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Secuencia de Aminoácidos , Animales , Sistemas CRISPR-Cas/genética , Línea Celular , Cromatografía Líquida de Alta Presión , Estrés del Retículo Endoplásmico/efectos de los fármacos , Edición Génica , Glicopéptidos/análisis , Glicosilación , Humanos , Ratones , Mutagénesis Sitio-Dirigida , N-Acetilglucosaminiltransferasas/deficiencia , N-Acetilglucosaminiltransferasas/genética , Receptor Notch1/genética , Receptor Notch1/metabolismo , Alineación de Secuencia , Espectrometría de Masas en Tándem , Tunicamicina/farmacología
14.
Methods Mol Biol ; 2132: 641-652, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32306363

RESUMEN

Many viruses utilize cell-surface glycans as receptors for host cell entry. Viral surface glycoproteins specifically interact with glycan motifs, which strongly contributes to viral tropism. Recently, the interactions between host cell glycan receptors and the mumps virus (MuV) hemagglutinin-neuraminidase (MuV-HN) protein were characterized by determining the co-crystal structure of MuV-HN in complex with glycan receptors. Here, we describe protocols for large-scale expression, purification and crystallization of MuV-HN proteins for structural analyses and glycan-binding assays with the overarching goal of investigating glycan-protein interactions.


Asunto(s)
Proteína HN/química , Proteína HN/metabolismo , Virus de la Parotiditis/fisiología , Paperas/virología , Polisacáridos/metabolismo , Cromatografía de Afinidad , Cromatografía en Gel , Cristalografía por Rayos X , Células HEK293 , Proteína HN/aislamiento & purificación , Humanos , N-Acetilglucosaminiltransferasas/deficiencia , Unión Proteica , Dominios Proteicos , Ingeniería de Proteínas , Tropismo Viral , Internalización del Virus
15.
FASEB J ; 34(6): 8385-8401, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32347583

RESUMEN

Certain proteoglycans, consisting of a core protein and glycosaminoglycan (GAG) chains, are among the many types of biomolecules that can function as damage-associated molecular pattern molecules (DAMPs). We, therefore, hypothesized that the expression level and structural alteration of GAGs affect inflammation. We have previously reported that the effects on GAG biosynthesis caused by loss of the tumor suppressor gene exostosin-like 2 (Extl2) influence liver injury and regeneration processes. To examine how altered GAG biosynthesis may underscore the relationship between inflammation and tumorigenesis, we assessed its role in non-alcoholic steatohepatitis and hepatocarcinoma (HCC) induced by dietary obesity and insulin-resistance. We demonstrated that GAGs produced in the absence of EXTL2 act as DAMPs and directly input signals into cells via the Toll-like 4 receptor. In addition, the subsequent transcriptional activation of inflammatory and tumor-promoting cytokines by NF-κB contributes to injury- and inflammation-driven tumor promotion. Thus, dysregulated biosynthesis of GAGs is considered to increase the risk of HCC in a background of obesity and diabetes.


Asunto(s)
Carcinogénesis/metabolismo , Glicosaminoglicanos/biosíntesis , Inflamación/metabolismo , Hígado/metabolismo , N-Acetilglucosaminiltransferasas/deficiencia , Transducción de Señal/fisiología , Receptor Toll-Like 4/metabolismo , Animales , Carcinogénesis/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Hepatitis/metabolismo , Hepatitis/patología , Inflamación/patología , Hígado/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Enfermedad del Hígado Graso no Alcohólico , Proteoglicanos/metabolismo , Células RAW 264.7
16.
Mucosal Immunol ; 13(5): 836-848, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32203062

RESUMEN

Modulation of immunity and disease by glycans is increasingly recognized. However, how host glycosylation shapes and is shaped by tuberculosis remains poorly understood. We show that deficiency in the glucosaminyl (N-acetyl) transferase 1 (Gcnt1), a key enzyme for core-2 O-glycans biosynthesis, drives susceptibility to Mycobacterium tuberculosis infection. The increased susceptibility of Gcnt1 deficient mice was characterized by extensive lung immune pathology, mechanistically related to neutrophils. Uninfected Gcnt1 deficient mice presented bone marrow, blood and lung neutrophilia, which further increased with infection. Blood neutrophilia required Gcnt1 deficiency in the hematopoietic compartment, relating with enhanced granulopoiesis, but normal cellular egress from the bone marrow. Interestingly, for the blood neutrophilia to translate into susceptibility to M. tuberculosis infection, Gnct1 deficiency in the stroma was also necessary. Complete Gcnt1 deficiency associated with increased lung expression of the neutrophil chemoattractant CXCL2. Lastly, we demonstrate that the transcript levels of various glycosyltransferase-encoding genes were altered in whole blood of active tuberculosis patients and that sialyl Lewis x, a glycan widely present in human neutrophils, was detected in the lung of tuberculosis patients. Our findings reveal a previously unappreciated link between Gcnt1, neutrophilia and susceptibility to M. tuberculosis infection, uncovering new players balancing the immune response in tuberculosis.


Asunto(s)
Predisposición Genética a la Enfermedad , Mycobacterium tuberculosis , N-Acetilglucosaminiltransferasas/deficiencia , Neutrófilos/inmunología , Neutrófilos/metabolismo , Tuberculosis/etiología , Tuberculosis/metabolismo , Animales , Carga Bacteriana , Biomarcadores , Modelos Animales de Enfermedad , Activación Enzimática , Regulación de la Expresión Génica , Glicosilación , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Ratones , Ratones Noqueados , Mycobacterium tuberculosis/inmunología , Neutrófilos/patología , Tasa de Supervivencia , Tuberculosis/diagnóstico , Tuberculosis/mortalidad
17.
Sci Rep ; 9(1): 12569, 2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31467334

RESUMEN

Small numbers of hematopoietic stem cells (HSCs) balance self-renewal and differentiation to produce the diversity and abundance of cell types that make up the blood system. How nutrients are recruited to support this massive differentiation and proliferation process remains largely unknown. The unique metabolism of adult HSCs, which rely on glycolysis and glutaminolysis, suggests a potential role for the post-translational modification O-GlcNAc as a critical nutrient signal in these cells. Glutamine, glucose, and other metabolites drive the hexosamine biosynthetic pathway (HBP) ultimately leading to the O-GlcNAc modification of critical intracellular targets. Here, we used a conditional targeted genetic deletion of the enzyme that removes O-GlcNAc, O-GlcNAcase (OGA), to determine the consequences of blocked O-GlcNAc cycling on HSCs. Oga deletion in mouse HSCs resulted in greatly diminished progenitor pools, impaired stem cell self-renewal and nearly complete loss of competitive repopulation capacity. Further, early T cell specification was particularly sensitive to Oga deletion. Loss of Oga resulted in a doubling of apoptotic cells within the bone marrow and transcriptional deregulation of key genes involved in adult stem cell maintenance and lineage specification. These findings suggest that O-GlcNAc cycling plays a critical role in supporting HSC homeostasis and early thymocyte development.


Asunto(s)
Acetilglucosamina/metabolismo , Células Madre Hematopoyéticas/citología , Linfocitos T/citología , Animales , Apoptosis , Diferenciación Celular , Autorrenovación de las Células , Femenino , Eliminación de Gen , Masculino , Ratones , N-Acetilglucosaminiltransferasas/deficiencia , N-Acetilglucosaminiltransferasas/genética , Especificidad de Órganos , Timocitos/citología , Transcripción Genética
18.
J Biol Chem ; 294(35): 13040-13050, 2019 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-31300553

RESUMEN

An early hallmark of type 2 diabetes is a failure of proinsulin-to-insulin processing in pancreatic ß-cells, resulting in hyperproinsulinemia. Proinsulin processing is quite sensitive to nutrient flux, and ß-cell-specific deletion of the nutrient-sensing protein modifier OGlcNAc transferase (ßOGTKO) causes ß-cell failure and diabetes, including early development of hyperproinsulinemia. The mechanisms underlying this latter defect are unknown. Here, using several approaches, including site-directed mutagenesis, Click O-GlcNAc labeling, immunoblotting, and immunofluorescence and EM imaging, we provide the first evidence for a relationship between the O-GlcNAcylation of eukaryotic translation initiation factor 4γ1 (eIF4G1) and carboxypeptidase E (CPE)-dependent proinsulin processing in ßOGTKO mice. We first established that ßOGTKO hyperproinsulinemia is independent of age, sex, glucose levels, and endoplasmic reticulum-CCAAT enhancer-binding protein homologous protein (CHOP)-mediated stress status. Of note, OGT loss was associated with a reduction in ß-cell-resident CPE, and genetic reconstitution of CPE in ßOGTKO islets rescued the dysfunctional proinsulin-to-insulin ratio. We show that although CPE is not directly OGlcNAc modified in islets, overexpression of the suspected OGT target eIF4G1, previously shown to regulate CPE translation in ß-cells, increases islet CPE levels, and fully reverses ßOGTKO islet-induced hyperproinsulinemia. Furthermore, our results reveal that OGT O-GlcNAc-modifies eIF4G1 at Ser-61 and that this modification is critical for eIF4G1 protein stability. Together, these results indicate a direct link between nutrient-sensitive OGT and insulin processing, underscoring the importance of post-translational O-GlcNAc modification in general cell physiology.


Asunto(s)
Carboxipeptidasa H/metabolismo , Diabetes Mellitus/metabolismo , Factor 4G Eucariótico de Iniciación/metabolismo , Células Secretoras de Insulina/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , N-Acetilglucosaminiltransferasas/deficiencia
19.
J Am Heart Assoc ; 8(11): e011260, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31131693

RESUMEN

Background Protein posttranslational modifications by O-linked ß-N-acetylglucosamine (O-GlcNAc) increase with cardiac hypertrophy, yet the functional effects of these changes are incompletely understood. In other organs, O-GlcNAc promotes adaptation to acute physiological stressors; however, prolonged O-GlcNAc elevations are believed to be detrimental. We hypothesize that early O-GlcNAcylation improves cardiac function during initial response to pressure overload hypertrophy, but that sustained elevations during established pathological hypertrophy negatively impact cardiac function by adversely affecting calcium handling proteins. Methods and Results Transverse aortic constriction or sham surgeries were performed on littermate controls or cardiac-specific, inducible O-GlcNAc transferase knockout (OGTKO) mice to reduce O-GlcNAc levels. O-GlcNAc transferase deficiency was induced at different times. To evaluate the initial response to pressure overload, OGTKO was completed preoperatively and mice were followed for 2 weeks post-surgery. To assess prolonged O-GlcNAcylation during established hypertrophy, OGTKO was performed starting 18 days after surgery and mice were followed until 6 weeks post-surgery. In both groups, OGTKO with transverse aortic constriction caused significant left ventricular dysfunction. OGTKO did not affect levels of the calcium handling protein SERCA2a. OGTKO reduced phosphorylation of phospholamban and cardiac troponin I, which would negatively impact cardiac function. O-GlcNAcylation of protein kinase A catalytic subunit, a kinase for phospholamban, decreased with OGTKO. Conclusions O-GlcNAcylation promotes compensated cardiac function in both early and established pathological hypertrophy. We identified a novel O-GlcNAcylation of protein kinase A catalytic subunit, which may regulate calcium handling and cardiac function.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hipertrofia Ventricular Izquierda/enzimología , Miocardio/enzimología , N-Acetilglucosaminiltransferasas/metabolismo , Disfunción Ventricular Izquierda/enzimología , Función Ventricular Izquierda , Remodelación Ventricular , Animales , Señalización del Calcio , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Glicosilación , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Ratones Noqueados , Miocardio/patología , N-Acetilglucosaminiltransferasas/deficiencia , N-Acetilglucosaminiltransferasas/genética , Fosforilación , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Factores de Tiempo , Troponina I/metabolismo , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
20.
Stem Cells ; 37(7): 899-909, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30977233

RESUMEN

As a morphogen, Sonic Hedgehog (Shh) mediates signaling at a distance from its sites of synthesis. After secretion, Shh must traverse a distance through the extracellular matrix (ECM) to reach the target cells and activate the Hh response. ECM proteins, in particular, the heparan sulfate proteoglycans (HSPGs) of the glypican family, have both negative and positive effects on Shh signaling, all attributed to their ability to bind Shh. Using mouse embryonic stem cell-derived mosaic tissues with compartments that lack the glycosyltransferases Exostosin1 and Exostosin2, or the HSPG core protein Glypican5, we show that Shh accumulates around its source cells when they are surrounded by cells that have a mutated ECM. This accumulation of Shh is correlated with an increased noncell autonomous Shh response. Our results support a model in which Shh presented on the cell surface accumulates at or near ECM that lacks HSPGs, possibly due to the absence of these Shh sequestering molecules. Stem Cells 2019;37:899-909.


Asunto(s)
Glipicanos/genética , Proteínas Hedgehog/genética , Células Madre Embrionarias de Ratones/metabolismo , N-Acetilglucosaminiltransferasas/genética , Transducción de Señal/genética , Animales , Diferenciación Celular , Linaje de la Célula/genética , Movimiento Celular , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Edición Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Glipicanos/deficiencia , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , N-Acetilglucosaminiltransferasas/deficiencia , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Transporte de Proteínas , Transfección , Proteína Fluorescente Roja
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA