Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 298(2): 101548, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34971707

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) continues to be a serious threat to the swine industry worldwide. Exostosin glycosyltransferase 1 (EXT1), an enzyme involved in the biosynthesis of heparin sulfate, has also been reported to be a host factor essential for a wide variety of pathogens. However, the role of EXT1 in PRRSV infection remains uncharted. Here, we identified that PRRSV infection caused an increase of EXT1 expression. EXT1 knockdown promoted virus infection, whereas its overexpression inhibited virus infection, suggesting an inhibitory function of EXT1 to PRRSV infection. We found that EXT1 had no effects on the attachment, internalization, or release of PRRSV but did restrict viral RNA replication. EXT1 was determined to interact with viral nonstructural protein 3 (nsp3) and nsp5 via its N-terminal cytoplasmic tail and to enhance K48-linked polyubiquitination of these two nsps to promote their degradation. Furthermore, the C-terminal glycosyltransferase activity domain of EXT1 was necessary for nsp3 and nsp5 degradation. We also found that EXT2, a EXT1 homolog, interacted with EXT1 and inhibited PRRSV infection. Similarly, EXT1 effectively restricted porcine epidemic diarrhea virus and porcine enteric alphacoronavirus infection in Vero cells. Taken together, this study reveals that EXT1 may serve as a broad-spectrum host restriction factor and suggests a molecular basis for the potential development of therapeutics against PRRSV infection.


Asunto(s)
N-Acetilglucosaminiltransferasas , Síndrome Respiratorio y de la Reproducción Porcina , Virus del Síndrome Respiratorio y Reproductivo Porcino , Proteínas no Estructurales Virales , Animales , Chlorocebus aethiops , N-Acetilglucosaminiltransferasas/metabolismo , Síndrome Respiratorio y de la Reproducción Porcina/enzimología , Síndrome Respiratorio y de la Reproducción Porcina/virología , Porcinos , Células Vero , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
2.
Am J Med Genet A ; 191(6): 1570-1575, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36869625

RESUMEN

Hereditary multiple exostoses (HME), also known as hereditary multiple osteochondroma (HMO), is an autosomal dominant disorder caused by pathogenic variants in exostosin-1 or -2 (EXT1 or EXT2). It is characterized by the formation of multiple benign growing osteochondromas (exostoses) that most commonly affect the long bones; however, it may also occur throughout the body. Although many of these lesions are clinically asymptomatic, some can lead to chronic pain and skeletal deformities and interfere with adjacent neurovascular structures. Here, we report two unrelated probands that presented with a clinical and molecular diagnosis of HME with venous malformation, a clinical feature not previously reported in individuals with HME.


Asunto(s)
Exostosis Múltiple Hereditaria , Humanos , Exostosis Múltiple Hereditaria/diagnóstico , Exostosis Múltiple Hereditaria/genética , N-Acetilglucosaminiltransferasas/genética , Mutación
3.
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
4.
Cytogenet Genome Res ; 162(1-2): 46-54, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35290978

RESUMEN

Langer-Giedion syndrome (LGS) is caused by a contiguous deletion at 8q23q24, characterized by exostoses, facial, ectodermal, and skeletal anomalies, and, occasionally, intellectual disability. LGS patients have been diagnosed clinically or by routine cytogenetic techniques, hampering the definition of an accurate genotype-phenotype correlation for the syndrome. We report two unrelated patients with 8q23q24 deletions, characterized by cytogenomic techniques, with one of them, to our knowledge, carrying the smallest deletion reported in classic LGS cases. We assessed the pathogenicity of the deletion of genes within the 8q23q24 region and reviewed other molecularly confirmed cases from the literature. Our findings suggest a 3.2-Mb critical region for a typical presentation of the syndrome, emphasizing the contribution of the TRPS1, RAD21, and EXT1 genes' haploinsufficiency, and facial dysmorphisms as well as bone anomalies as the most frequent features among patients with LGS. We also suggest a possible role for the CSMD3 gene, whose deletion seems to contribute to central nervous system anomalies. Since studies performing such correlation for LGS patients are limited, our data contribute to improving the ge-notype-phenotype characterization for LGS patients.


Asunto(s)
Síndrome de Langer-Giedion , Deleción Cromosómica , Cromosomas Humanos Par 8 , Hibridación Genómica Comparativa , Estudios de Asociación Genética , Haploinsuficiencia , Humanos , Síndrome de Langer-Giedion/diagnóstico , Síndrome de Langer-Giedion/genética , Fenotipo , Proteínas Represoras/genética
5.
Medicina (Kaunas) ; 59(1)2022 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-36676722

RESUMEN

Background and Objectives: Hereditary multiple exostoses (HME) is a disease characterized by cartilage-capped bony protuberances at the site of growth plates of long bones. Functional mutations in the exostosin genes (EXT1 and EXT2) are reported to affect the hedgehog signalling pathways leading to multiple enchondromatosis. However, the exact role of each EXT protein in the regulation of heparan sulphate (HS) chain elongation is still an enigma. In this study, a Pakistani family with HME is investigated to find out the genetic basis of the disease. Materials and Methods: Genotyping of eight members of the family by amplifying microsatellite markers, tightly linked to the EXT1 and EXT2 genes. Results: The study revealed linkage of the HME family to the EXT1 locus 8q24.1. Sanger sequencing identified a heterozygous deletion (c.247Cdel) in exon 1 of EXT1, segregating with the disease phenotype in the family. In silico analysis predicted a shift in the frame causing an early stop codon (p.R83GfsX52). The predicted dwarf protein constituting 134 amino acids was functionally aberrant with a complete loss of the catalytic domain at the C-terminus. Interestingly, an alternative open reading frame 3 (ORF3) caused by the frame shift is predicted to encode a protein sequence, identical to the wild type and containing the catalytic domain, but lacking the first 100 amino acids of the wild-type EXT1 protein. Conclusion: Consequently, haploinsufficiency could be the cause of HME in the investigated family as the mutated copy of EXT1 is ineffective for EXT-1/2 complex formation. The predicted ORF3 protein could be of great significance in understanding several aspects of HME pathogenesis.


Asunto(s)
Exostosis Múltiple Hereditaria , Humanos , Exostosis Múltiple Hereditaria/genética , Exostosis Múltiple Hereditaria/patología , Haploinsuficiencia/genética , Pakistán , Proteínas Hedgehog/genética , Mutación , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , Heparitina Sulfato/metabolismo , Aminoácidos/genética
6.
J Cell Mol Med ; 25(5): 2609-2620, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33565239

RESUMEN

DNA methylation is important for lung cancer prognosis. In this work, it is aimed to seek novel biomarkers with DNA methylation-expression-pathway pattern and explore its underlying mechanism. Prognostic DNA methylation sites and mRNAs were screened in NSCLC data set from TCGA, and further validated using the samples retrospectively collected, and EXT1 was identified as a potential target. Gene body methylation of three CpG sites (cg03276982, cg11592677, cg16286281) on EXT1 was significantly associated with clinical outcome, and the EXT1 gene expression also predicted prognosis. The expression level of EXT1 was also correlated with its DNA methylation level. This observation was further validated in a new data set consist of 170 samples. Knocking down of EXT1 resulted in decreased proliferation and migration. EXT1 targets were analysed using GSEA. It is found that the WNT signalling is the potential downstream target of EXT1. Further analyses revealed that the EXT1 targets the beta-catenin and effect migration rate of NSCLC cell lines. The WNT signalling inhibitor, XAV-939, effectively disrupted the migration promotion effect induced by EXT1. In summary, EXT1 methylation regulates the gene expression, effects the proliferation and migration via WNT pathway and predicted a poor prognosis for NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , N-Acetilglucosaminiltransferasas/genética , Vía de Señalización Wnt , Biomarcadores de Tumor , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Islas de CpG , Humanos , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Análisis de Supervivencia
7.
Am J Kidney Dis ; 77(3): 440-453, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33487481

RESUMEN

The understanding and management of membranous nephropathy, a common cause of nephrotic syndrome that is more frequently encountered in adults than in children, has rapidly evolved over the past decade. Identification of target antigens has allowed for more precise molecular diagnoses, and the ability to monitor circulating autoantibodies has added a new vantage point in terms of disease monitoring and decisions about immunosuppression. Although immunosuppression with alkylating agents combined with corticosteroids, or with calcineurin inhibitor-based regimens, has been the historical mainstay of treatment, observational and now randomized controlled trials with the B-cell-depleting agent rituximab have moved this agent to the forefront of therapy for primary membranous nephropathy. In this Core Curriculum, we discuss the typical features of primary and secondary disease; highlight the target antigens such as the phospholipase A2 receptor, thrombospondin type 1 domain-containing 7A, neural epidermal growth factor-like 1, and semaphorin-3B; describe the relationship between the immunologic and clinical courses of disease; and review modern management with supportive care or immunosuppressive treatment based on these composite parameters.


Asunto(s)
Autoanticuerpos/inmunología , Glomerulonefritis Membranosa/inmunología , Proteínas de Unión al Calcio/inmunología , Glomerulonefritis Membranosa/tratamiento farmacológico , Glomerulonefritis Membranosa/etiología , Glomerulonefritis Membranosa/patología , Glucocorticoides/uso terapéutico , Enfermedad Injerto contra Huésped/complicaciones , Enfermedad Injerto contra Huésped/inmunología , Hepatitis B/complicaciones , Hepatitis C/complicaciones , Humanos , Factores Inmunológicos/uso terapéutico , Inmunosupresores , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/inmunología , Malaria/complicaciones , Glicoproteínas de Membrana/inmunología , Enfermedad Mixta del Tejido Conjuntivo/complicaciones , Enfermedad Mixta del Tejido Conjuntivo/inmunología , Neoplasias/complicaciones , Receptores de Fosfolipasa A2/inmunología , Rituximab/uso terapéutico , Semaforinas/inmunología , Síndrome de Sjögren/complicaciones , Síndrome de Sjögren/inmunología , Trombospondinas/inmunología
8.
Respir Res ; 22(1): 193, 2021 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-34217286

RESUMEN

BACKGROUND: Endothelial glycocalyx loss is integral to increased pulmonary vascular permeability in sepsis-related acute lung injury. Protectin conjugates in tissue regeneration 1 (PCTR1) is a novel macrophage-derived lipid mediator exhibiting potential anti-inflammatory and pro-resolving benefits. METHODS: PCTR1 was administrated intraperitoneally with 100 ng/mouse after lipopolysaccharide (LPS) challenged. Survival rate and lung function were used to evaluate the protective effects of PCTR1. Lung inflammation response was observed by morphology and inflammatory cytokines level. Endothelial glycocalyx and its related key enzymes were measured by immunofluorescence, ELISA, and Western blot. Afterward, related-pathways inhibitors were used to identify the mechanism of endothelial glycocalyx response to PCTR1 in mice and human umbilical vein endothelial cells (HUVECs) after LPS administration. RESULTS: In vivo, we show that PCTR1 protects mice against lipopolysaccharide (LPS)-induced sepsis, as shown by enhanced the survival and pulmonary function, decreased the inflammatory response in lungs and peripheral levels of inflammatory cytokines such as tumor necrosis factor-α, interleukin-6, and interleukin-1ß. Moreover, PCTR1 restored lung vascular glycocalyx and reduced serum heparin sulphate (HS), syndecan-1 (SDC-1), and hyaluronic acid (HA) levels. Furthermore, we found that PCTR1 downregulated heparanase (HPA) expression to inhibit glycocalyx degradation and upregulated exostosin-1 (EXT-1) protein expression to promote glycocalyx reconstitution. Besides, we observed that BAY11-7082 blocked glycocalyx loss induced by LPS in vivo and in vitro, and BOC-2 (ALX antagonist) or EX527 (SIRT1 inhibitor) abolished the restoration of HS in response to PCTR1. CONCLUSION: PCTR1 protects endothelial glycocalyx via ALX receptor by regulating SIRT1/NF-κB pathway, suggesting PCTR1 may be a significant therapeutic target for sepsis-related acute lung injury.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antiinflamatorios/farmacología , Glicocálix/metabolismo , FN-kappa B/metabolismo , Mucosa Respiratoria/metabolismo , Sirtuina 1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Animales , Ácidos Docosahexaenoicos/farmacología , Glicocálix/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Lipopolisacáridos/toxicidad , Masculino , Ratones , FN-kappa B/antagonistas & inhibidores , Mucosa Respiratoria/efectos de los fármacos , Sirtuina 1/antagonistas & inhibidores
9.
Hum Genomics ; 14(1): 18, 2020 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-32398079

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease affecting the central nervous system in young adults. Heparan sulfate proteoglycans (HSPGs) are ubiquitous to the cell surface and the extracellular matrix. HSPG biosynthesis is a complex process involving enzymatic attachment of heparan sulfate (HS) chains to a core protein. HS side chains mediate specific ligand and growth factor interactions directing cellular processes including cell adhesion, migration and differentiation. Two main families of HSPGs exist, the syndecans (SDC1-4) and glypicans (GPC1-6). The SDCs are transmembrane proteins, while the GPC family are GPI linked to the cell surface. SDC1 has well-documented interactions with numerous signalling pathways. Genome-wide association studies (GWAS) have identified regions of the genome associated with MS including a region on chromosome 13 containing GPC5 and GPC6. International studies have revealed significant associations between this region and disease development. The exostosin-1 (EXT1) and sulfatase-1 (SULF1) are key enzymes contributing to the generation of HS chains. EXT1, with documented tumour suppressor properties, is involved in the initiation and polymerisation of the growing HS chain. SULF1 removes 6-O-sulfate groups from HS chains, affecting protein-ligand interactions and subsequent downstream signalling with HS modification potentially having significant effects on MS progression. In this study, we identified significant associations between single nucleotide polymorphisms in SDC1, GPC5 and GPC6 and MS in an Australian Caucasian case-control population. Further significant associations in these genes were identified when the population was stratified by sex and disease subtype. No association was found for EXT1 or SULF1.


Asunto(s)
Biomarcadores/análisis , Estudio de Asociación del Genoma Completo , Proteoglicanos de Heparán Sulfato/química , Esclerosis Múltiple/patología , Polimorfismo de Nucleótido Simple , Población Blanca/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Australia/epidemiología , Estudios de Casos y Controles , Femenino , Glipicanos/genética , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/enzimología , Esclerosis Múltiple/epidemiología , Esclerosis Múltiple/genética , N-Acetilglucosaminiltransferasas/genética , Sulfotransferasas/genética , Sindecano-1/genética , Adulto Joven
10.
J Cell Sci ; 131(10)2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29700203

RESUMEN

Heparan sulfate proteoglycans (HSPGs) have been shown to regulate various developmental processes. However, the function of heparan sulfate (HS) during the development of mammalian stomach has not been characterized yet. Here, we investigate the role of epithelial HS in embryonic stomach by examining mice deficient in the glycosyltransferase gene Ext1 We show that HS exhibits a specific and dynamic expression pattern in mouse embryonic stomach. Depletion of the epithelial HS leads to stomach hypoplasia, with phenotypic differences in the gastric mucosa between the forestomach and hindstomach. In the posterior stomach, HS depletion disrupts glandular stomach patterning and cytodifferentiation via attenuation of Fgf signaling activity. Inhibition of Fgf signaling in vitro recapitulates the patterning defect. Ligand and carbohydrate engagement assay (LACE) reveals a diminished assembly of Fgf10 and Fgfr2b in the mutant. In the anterior stomach, loss of epithelial HS leads to stratification and differentiation defects of the multilayered squamous epithelium, along with reduced Hh and Bmp signaling activity. Our data demonstrate that epithelial HS plays multiple roles in regulating mammalian stomach morphogenesis in a regional-specific manner.


Asunto(s)
Epitelio/metabolismo , Heparitina Sulfato/metabolismo , Estómago/embriología , Animales , Epitelio/embriología , Femenino , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Mucosa Gástrica/metabolismo , Masculino , Ratones , Ratones Noqueados , Morfogénesis , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo
11.
Osteoarthritis Cartilage ; 28(11): 1459-1470, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32818603

RESUMEN

OBJECTIVE: Exostosin-1 (EXT1) and EXT2 are the major genetic etiologies of multiple hereditary exostoses and are essential for heparan sulfate (HS) biosynthesis. Previous studies investigating HS in several mouse models of multiple hereditary exostoses have reported that aberrant bone morphogenetic protein (BMP) signaling promotes osteochondroma formation in Ext1-deficient mice. This study examined the mechanism underlying the effects of HS deficiency on BMP/Smad signaling in articular cartilage in a cartilage-specific Ext-/- mouse model. METHOD: We generated mice with a conditional Ext1 knockout in cartilage tissue (Ext1-cKO mice) using Prg4-Cre transgenic mice. Structural cartilage alterations were histologically evaluated and phospho-Smad1/5/9 (pSmad1/5/9) expression in mouse chondrocytes was analyzed. The effect of pharmacological intervention of BMP signaling using a specific inhibitor was assessed in the articular cartilage of Ext1-cKO mice. RESULTS: Hypertrophic chondrocytes were significantly more abundant (P = 0.021) and cartilage thickness was greater in Ext1-cKO mice at 3 months postnatal than in control littermates (P = 0.036 for femur; and P < 0.001 for tibia). However, osteoarthritis did not spontaneously occur before the 1-year follow-up. matrix metalloproteinase (MMP)-13 and adamalysin-like metalloproteinases with thrombospondin motifs(ADAMTS)-5 were upregulated in hypertrophic chondrocytes of transgenic mice. Immunostaining and western blotting revealed that pSmad1/5/9-positive chondrocytes were more abundant in the articular cartilage of Ext1-cKO mice than in control littermates. Furthermore, the BMP inhibitor significantly decreased the number of hypertrophic chondrocytes in Ext1-cKO mice (P = 0.007). CONCLUSIONS: HS deficiency in articular chondrocytes causes chondrocyte hypertrophy, wherein upregulated BMP/Smad signaling partially contributes to this phenotype. HS might play an important role in maintaining the cartilaginous matrix by regulating BMP signaling.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Heparitina Sulfato/deficiencia , Osteoartritis de la Rodilla/metabolismo , Proteína ADAMTS5/metabolismo , Animales , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Cartílago Articular/citología , Condrocitos/patología , Modelos Animales de Enfermedad , Hipertrofia , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , N-Acetilglucosaminiltransferasas/genética , Osteoartritis de la Rodilla/genética , Osteoartritis de la Rodilla/patología , Pirazoles/farmacología , Pirimidinas/farmacología , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo
12.
Osteoarthritis Cartilage ; 28(7): 977-987, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32315715

RESUMEN

OBJECTIVE: Osteoarthritis (OA) is a progressive degenerative disease of the articular cartilage caused by an unbalanced activity of proteases, cytokines and other secreted proteins. Since heparan sulfate (HS) determines the activity of many extracellular factors, we investigated its role in OA progression. METHODS: To analyze the role of the HS level, OA was induced by anterior cruciate ligament transection (ACLT) in transgenic mice carrying a loss-of-function allele of Ext1 in clones of chondrocytes (Col2-rtTA-Cre;Ext1e2fl/e2fl). To study the impact of the HS sulfation pattern, OA was surgically induced in mice with a heterozygous (Ndst1+/-) or chondrocyte-specific (Col2-Cre;Ndst1fl/fl) loss-of-function allele of the sulfotransferase Ndst1. OA progression was evaluated using the OARSI scoring system. To investigate expression and activity of cartilage degrading proteases, femoral head explants of Ndst1+/- mutants were analyzed by qRT-PCR, Western Blot and gelatin zymography. RESULTS: All investigated mouse strains showed reduced OA scores (Col2-rtTA-Cre;Ext1e2fl/e2fl: 0.83; 95% HDI 0.72-0.96; Ndst1+/-: 0.83, 95% HDI 0.74-0.9; Col2-Cre;Ndst1fl/fl: 0.87, 95% HDI 0.76-1). Using cartilage explant cultures of Ndst1 animals, we detected higher amounts of aggrecan degradation products in wildtype samples (NITEGE 4.24-fold, 95% HDI 1.05-18.55; VDIPEN 1.54-fold, 95% HDI 1.54-2.34). Accordingly, gelatin zymography revealed lower Mmp2 activity in mutant samples upon RA-treatment (0.77-fold, 95% HDI: 0.60-0.96). As expression of major proteases and their inhibitors was not altered, HS seems to regulate cartilage degeneration by affecting protease activity. CONCLUSION: A decreased HS content or a reduced sulfation level protect against OA progression by regulating protease activity rather than expression.


Asunto(s)
Cartílago Articular/metabolismo , Condrocitos/metabolismo , Heparitina Sulfato/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Osteoartritis/metabolismo , Agrecanos/metabolismo , Animales , Ligamento Cruzado Anterior/cirugía , Western Blotting , Cartílago Articular/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Mutación con Pérdida de Función , Ratones , Ratones Transgénicos , N-Acetilglucosaminiltransferasas/genética , Osteoartritis/genética , Osteoartritis/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Sulfotransferasas/genética
13.
Osteoporos Int ; 31(12): 2355-2361, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32642853

RESUMEN

We analyzed osteoporosis in 20 HME patients. According to the T-score of BMD, 30% and 67.5% of the patients fell in the range of osteopenia in the lumbar spine and femoral neck. Our results indicate HME patients have low bone mass. They do not have abnormal bone metabolism. INTRODUCTION: There are few reports of osteoporosis in hereditary multiple exostoses (HME) patients. Therefore, the purpose of this study was to analyze osteoporosis in HME patients. METHODS: This retrospective cohort study included 20 patients diagnosed with HME. Patients underwent bone mineral density (BMD) measurement of the lumbar spine (n = 20) and femoral neck (n = 40). Bone metabolic parameters, including serum osteocalcin and urinary cross-linked N-telopeptide of type 1 collagen (NTx), were analyzed in all subjects. EXT1 and EXT2 genes were sequenced using genomic DNA. We also examined the correlation between genotype and BMD Z-score and T-score. RESULTS: The mean BMD values of the lumbar spine were 1.085 ± 0.116 g/cm2 (n = 11) in male and 1.108 ± 0.088 g/cm2 (n = 9) in female. The mean BMD values of the femoral neck area were 0.759 ± 0.125 g/cm2 (n = 22) in male and 0.749 ± 0.115 g/cm2 (n = 18) in female. Z-score of most HME patients show < 0, indicating that these patients tend to have low bone mass compared with the age-matched population. According to the T-score of BMD, 30% (6 of 20) and 67.5% (27 of 40) of the patients fell in the range of osteopenia in the lumbar spine and femoral neck areas, respectively. Serum osteocalcin and urinary NTx were in the normal range in most patients. There was no significant correlation between genotypes and Z-score. CONCLUSION: HME patients have low bone mass, especially in the femoral neck area. They do not have abnormal bone metabolism, and there was no correlation between genotypes and Z-score.


Asunto(s)
Exostosis Múltiple Hereditaria , Osteoporosis , Densidad Ósea , Femenino , Cuello Femoral , Humanos , Vértebras Lumbares , Masculino , Osteoporosis/epidemiología , Osteoporosis/etiología , Estudios Retrospectivos
14.
Osteoarthritis Cartilage ; 27(11): 1702-1710, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31330188

RESUMEN

OBJECTIVE: Exostosin-1 (Ext1) encodes a glycosyltransferase required for heparan sulfate (HS) chain elongation in HS-proteoglycan biosynthesis. HS chains serve as binding partners for signaling proteins, affecting their distribution and activity. The Wnt/ß-catenin pathway emerged as critical regulator of chondrogenesis. Yet, how EXT1 and HS affect Wnt/ß-catenin signaling during chondrogenesis remains unexplored. METHOD: Ext1 was stably knocked-down or overexpressed in ATDC5 chondrogenic cells cultured as micromasses. HS content was determined using ELISA. Chondrogenic markers Sox9, Col2a1, Aggrecan, and Wnt direct target gene Axin2 were measured by RT-qPCR. Proteoglycan content was evaluated by Alcian blue and DMMB assay, canonical Wnt signaling activation by ß-catenin Western blot and TOP/FOP assay. ATDC5 cells and human articular chondrocytes were treated with Wnt activators CHIR99021 and recombinant WNT3A. RESULTS: Ext1 knock-down reduced HS, and increased chondrogenic markers and proteoglycan accumulation. Ext1 knock-down reduced active Wnt/ß-catenin signaling. Conversely, Ext1 overexpressing cells, with higher HS content, showed decreased chondrogenic differentiation and enhanced Wnt/ß-catenin signaling. Wnt/ß-catenin signaling activation led to a down-regulation of Ext1 expression in ATDC5 cells and in human articular chondrocytes. CONCLUSIONS: EXT1 affects chondrogenic differentiation of precursor cells, in part via changes in the activity of Wnt/ß-catenin signaling. Wnt/ß-catenin signaling controls Ext1 expression, suggesting a regulatory loop between EXT1 and Wnt/ß-catenin signaling during chondrogenesis.


Asunto(s)
Condrocitos/metabolismo , Condrogénesis/genética , Regulación de la Expresión Génica , N-Acetilglucosaminiltransferasas/genética , ARN/genética , Vía de Señalización Wnt/genética , Western Blotting , Diferenciación Celular , Células Cultivadas , Condrocitos/patología , Ensayo de Inmunoadsorción Enzimática , Humanos , N-Acetilglucosaminiltransferasas/biosíntesis , Proteínas Wnt/biosíntesis , Proteínas Wnt/genética
15.
Biochem Biophys Res Commun ; 505(4): 959-965, 2018 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-30262140

RESUMEN

OBJECTIVE: Screening and identifying the gene mutation of EXT1, EXT2 and EXT3 associated with multiple exostosis (ME) and the expression in tumor tissues. METHODS: Nine patients with multiple exostosis were collected and genomic DNA was extracted. Polymerase chain reaction (PCR) amplification and direct sequencing techniques were used to screen all exons, 5' and 3' ends of the EXT1, EXT2 and EXT3 related causative genes. EXT1, EXT2 and EXT3 gene were screened and quantified by RNA-SEQ and RT-qPCR. The concentration of calcitonin gene-related peptide (CGRP) in peripheral blood of tumor patients and normal controls was detected by ELISA. RESULTS: Between the two patients with ME, the EXT1 gene was found in one patient to have c.79 T>A mutation, which caused the change of p.M27T, the non polar methionine was replaced by the high frequency mutation of polar threonine, and the rest of patients was found the splicing mutation c.1284 + 8 delAT of the heterozygosity of the EXT1 gene. The serum CGRP concentration of ME patients (623 + 49 pg/ml) was significantly higher than that of normal controls (196 + 68 pg/ml), and EXT1 mutation patients were also higher than non mutation patients.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/genética , Exostosis Múltiple Hereditaria/genética , N-Acetilglucosaminiltransferasas/genética , Péptido Relacionado con Gen de Calcitonina/sangre , Biología Computacional , Exostosis Múltiple Hereditaria/diagnóstico , Perfilación de la Expresión Génica , Humanos , Mutación
16.
Connect Tissue Res ; 59(1): 85-98, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29099240

RESUMEN

Multiple hereditary exostoses (MHE) is an autosomal dominant disorder that affects about 1 in 50,000 children worldwide. MHE, also known as hereditary multiple exostoses (HME) or multiple osteochondromas (MO), is characterized by cartilage-capped outgrowths called osteochondromas that develop adjacent to the growth plates of skeletal elements in young patients. These benign tumors can affect growth plate function, leading to skeletal growth retardation, or deformations, and can encroach on nerves, tendons, muscles, and other surrounding tissues and cause motion impairment, chronic pain, and early onset osteoarthritis. In about 2-5% of patients, the osteochondromas can become malignant and life threatening. Current treatments consist of surgical removal of the most symptomatic tumors and correction of the major skeletal defects, but physical difficulties and chronic pain usually continue and patients may undergo multiple surgeries throughout life. Thus, there is an urgent need to find new treatments to prevent or reverse osteochondroma formation. The 2016 International MHE Research Conference was convened to provide a forum for the presentation of the most up-to-date and advanced clinical and basic science data and insights in MHE and related fields; to stimulate the forging of new perspectives, collaborations, and venues of research; and to publicize key scientific findings within the biomedical research community and share insights and relevant information with MHE patients and their families. This report provides a description, review, and assessment of all the exciting and promising studies presented at the Conference and delineates a general roadmap for future MHE research targets and goals.


Asunto(s)
Exostosis Múltiple Hereditaria , Animales , Congresos como Asunto , Exostosis Múltiple Hereditaria/metabolismo , Exostosis Múltiple Hereditaria/patología , Exostosis Múltiple Hereditaria/terapia , Humanos
17.
Biochim Biophys Acta Gen Subj ; 1862(6): 1472-1481, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29580921

RESUMEN

BACKGROUND: Exostosin-1 (EXT1), a member of the EXT protein family, is indispensable for synthesis of heparan sulfate (HS) chains that bind to and modulate the signaling efficiency of numerous growth factor activities. We have previously shown that Ext1 mutated mouse embryonic fibroblasts produce short sulfated HS chains which dramatically influence tumor cell behavior in a 3-dimensional (3D) heterospheroid system composed of tumor cells and fibroblasts. METHODS: In this study, we have used both 2D co-culture and 3D heterospheroid models, consisting of human A549 carcinoma cells co-cultured with wild-type or Ext1-mutated mouse embryonic fibroblasts. RESULTS AND CONCLUSIONS: Gene expression profiling of differentially expressed genes in fibroblast/A549 heterospheroids identified P311 as a gene substantially down-regulated in A549 cells co-cultured with Ext1-mutated fibroblasts. In addition, we observed that the Ext1 mutants displayed reduced Tgf-ß1 mRNA levels and lower levels of secreted active TGF-ß protein. Re-introduction of Ext1 in the Ext1 mutant fibroblasts rescued the levels of Tgf-ß1 mRNA, increased the amounts of secreted active TGF-ß in these cells, as well as P311 mRNA levels in adjacent A549 cells. Accordingly, small interfering RNAs (siRNAs) against fibroblast Tgf-ß1 reduced P311 expression in neighboring A549 tumor cells. Our data raises the possibility that fibroblast Ext1 levels play a role in P311 expression in A549/fibroblast co-culture through TGF-ß1. GENERAL SIGNIFICANCE: This study considers a possible novel mechanism of Ext1-regulated heparan sulfate structure in modifying tumor-stroma interactions through altering stromal tgf-ß1 expression.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Heparitina Sulfato/farmacología , N-Acetilglucosaminiltransferasas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células A549 , Animales , Técnicas de Cocultivo , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Humanos , Ratones , N-Acetilglucosaminiltransferasas/genética , Proteínas del Tejido Nervioso/genética , Factor de Crecimiento Transformador beta1/metabolismo
18.
Glycoconj J ; 34(3): 411-420, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27744520

RESUMEN

Proteoglycans and glycosaminoglycans modulate numerous cellular processes relevant to tumour progression, including cell proliferation, cell-matrix interactions, cell motility and invasive growth. Among the glycosaminoglycans with a well-documented role in tumour progression are heparan sulphate, chondroitin/dermatan sulphate and hyaluronic acid/hyaluronan. While the mode of biosynthesis differs for sulphated glycosaminoglycans, which are synthesised in the ER and Golgi compartments, and hyaluronan, which is synthesized at the plasma membrane, these polysaccharides partially compete for common substrates. In this study, we employed a siRNA knockdown approach for heparan sulphate (EXT1) and heparan/chondroitin/dermatan sulphate-biosynthetic enzymes (ß4GalT7) in the aggressive human breast cancer cell line MDA-MB-231 to study the impact on cell behaviour and hyaluronan biosynthesis. Knockdown of ß4GalT7 expression resulted in a decrease in cell viability, motility and adhesion to fibronectin, while these parameters were unchanged in EXT1-silenced cells. Importantly, these changes were associated with a decreased expression of syndecan-1, decreased signalling response to HGF and an increase in the synthesis of hyaluronan, due to an upregulation of the hyaluronan synthases HAS2 and HAS3. Interestingly, EXT1-depleted cells showed a downregulation of the UDP-sugar transporter SLC35D1, whereas SLC35D2 was downregulated in ß4GalT7-depleted cells, indicating an intricate regulatory network that connects all glycosaminoglycans synthesis. The results of our in vitro study suggest that a modulation of breast cancer cell behaviour via interference with heparan sulphate biosynthesis may result in a compensatory upregulation of hyaluronan biosynthesis. These findings have important implications for the development of glycosaminoglycan-targeted therapeutic approaches for malignant diseases.


Asunto(s)
Sulfatos de Condroitina/biosíntesis , Dermatán Sulfato/análogos & derivados , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Heparitina Sulfato/biosíntesis , Ácido Hialurónico/biosíntesis , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Sulfatos de Condroitina/antagonistas & inhibidores , Sulfatos de Condroitina/genética , Dermatán Sulfato/antagonistas & inhibidores , Dermatán Sulfato/biosíntesis , Dermatán Sulfato/genética , Células Epiteliales/patología , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Heparitina Sulfato/antagonistas & inhibidores , Heparitina Sulfato/genética , Humanos , Hialuronano Sintasas/antagonistas & inhibidores , Hialuronano Sintasas/genética , Hialuronano Sintasas/metabolismo , Ácido Hialurónico/antagonistas & inhibidores , Ácido Hialurónico/genética , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Proteínas de Transporte de Monosacáridos/antagonistas & inhibidores , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , N-Acetilglucosaminiltransferasas/antagonistas & inhibidores , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , N-Acetil-Lactosamina Sintasa/antagonistas & inhibidores , N-Acetil-Lactosamina Sintasa/genética , N-Acetil-Lactosamina Sintasa/metabolismo , Proteínas de Transporte de Nucleótidos/antagonistas & inhibidores , Proteínas de Transporte de Nucleótidos/genética , Proteínas de Transporte de Nucleótidos/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal
19.
Pediatr Blood Cancer ; 64(3)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27667142

RESUMEN

Transient myeloproliferative disorder (TMD) is a leukemia type that occurs typically in newborns. In Down syndrome, TMD is referred to as transient abnormal myelopoiesis (TAM).32 Recently, transientness has also been reported in acute myeloid leukemia patients with germline trisomy 21 mosaicism, and even in cases with somatic trisomy 21, with or without GATA1 mutations. TMD cases without trisomy 21 are rare, and recurrent genetic aberrations that aid in clinical decision-making are scarcely described. We describe here a TMD patient without trisomy 21 or GATA1 mutation in whom single-nucleotide polymorphism analysis of leukemic blasts revealed a novel combined submicroscopic deletion (5q31.1-5q31.3 and 8q23.2q24).


Asunto(s)
Cromosomas Humanos Par 5/genética , Cromosomas Humanos Par 8/genética , Síndrome de Down/genética , Factor de Transcripción GATA1/genética , Leucemia Megacarioblástica Aguda/genética , Polimorfismo de Nucleótido Simple/genética , Síndrome de Down/patología , Humanos , Recién Nacido , Leucemia Megacarioblástica Aguda/patología , Pronóstico
20.
Curr Osteoporos Rep ; 15(3): 142-152, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28466453

RESUMEN

PURPOSE OF REVIEW: Hereditary multiple exostoses (HME) is a complex musculoskeletal pediatric disorder characterized by osteochondromas that form next to the growth plates of many skeletal elements, including long bones, ribs, and vertebrae. Due to its intricacies and unresolved issues, HME continues to pose major challenges to both clinicians and biomedical researchers. The purpose of this review is to describe and analyze recent advances in this field and point to possible targets and strategies for future biologically based therapeutic intervention. RECENT FINDINGS: Most HME cases are linked to loss-of-function mutations in EXT1 or EXT2 that encode glycosyltransferases responsible for heparan sulfate (HS) synthesis, leading to HS deficiency. Recent genomic inquiries have extended those findings but have yet to provide a definitive genotype-phenotype correlation. Clinical studies emphasize that in addition to the well-known skeletal problems caused by osteochondromas, HME patients can experience, and suffer from, other symptoms and health complications such as chronic pain and nerve impingement. Laboratory work has produced novel insights into alterations in cellular and molecular mechanisms instigated by HS deficiency and subtending onset and growth of osteochondroma and how such changes could be targeted toward therapeutic ends. HME is a rare and orphan disease and, as such, is being studied only by a handful of clinical and basic investigators. Despite this limitation, significant advances have been made in the last few years, and the future bodes well for deciphering more thoroughly its pathogenesis and, in turn, identifying the most effective treatment for osteochondroma prevention.


Asunto(s)
Exostosis Múltiple Hereditaria/fisiopatología , Dolor Crónico/etiología , Exostosis Múltiple Hereditaria/complicaciones , Exostosis Múltiple Hereditaria/genética , Humanos , Mutación , N-Acetilglucosaminiltransferasas/genética , Síndromes de Compresión Nerviosa/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA