Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
FASEB J ; 33(2): 2669-2679, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30307768

RESUMO

Congenital disorders of glycosylation are severe inherited diseases in which aberrant protein glycosylation is a hallmark. Transmembrane protein 165 (TMEM165) is a novel Golgi transmembrane protein involved in type II congenital disorders of glycosylation. Although its biologic function is still a controversial issue, we have demonstrated that the Golgi glycosylation defect due to TMEM165 deficiency resulted from a Golgi Mn2+ homeostasis defect. The goal of this study was to delineate the cellular pathway by which extracellular Mn2+ rescues N-glycosylation in TMEM165 knockout (KO) cells. We first demonstrated that after extracellular exposure, Mn2+ uptake by HEK293 cells at the plasma membrane did not rely on endocytosis but was likely done by plasma membrane transporters. Second, we showed that the secretory pathway Ca2+-ATPase 1, also known to mediate the influx of cytosolic Mn2+ into the lumen of the Golgi apparatus, is not crucial for the Mn2+-induced rescue glycosylation of lysosomal-associated membrane protein 2 (LAMP2). In contrast, our results demonstrate the involvement of cyclopiazonic acid- and thapsigargin (Tg)-sensitive pumps in the rescue of TMEM165-associated glycosylation defects by Mn2+. Interestingly, overexpression of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) 2b isoform in TMEM165 KO cells partially rescues the observed LAMP2 glycosylation defect. Overall, this study indicates that the rescue of Golgi N-glycosylation defects in TMEM165 KO cells by extracellular Mn2+ involves the activity of Tg and cyclopiazonic acid-sensitive pumps, probably the SERCA pumps.-Houdou, M., Lebredonchel, E., Garat, A., Duvet, S., Legrand, D., Decool, V., Klein, A., Ouzzine, M., Gasnier, B., Potelle, S., Foulquier, F. Involvement of thapsigargin- and cyclopiazonic acid-sensitive pumps in the rescue of TMEM165-associated glycosylation defects by Mn2+.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Indóis/farmacologia , Manganês/farmacologia , Proteínas de Membrana/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Tapsigargina/farmacologia , Antiporters , Transporte Biológico , Cálcio/metabolismo , Proteínas de Transporte de Cátions , Defeitos Congênitos da Glicosilação/tratamento farmacológico , Defeitos Congênitos da Glicosilação/metabolismo , Defeitos Congênitos da Glicosilação/patologia , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Inibidores Enzimáticos/farmacologia , Glicosilação , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Células HEK293 , Homeostase , Humanos , Proteína 2 de Membrana Associada ao Lisossomo/genética , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética
2.
J Biol Chem ; 290(12): 7658-70, 2015 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-25568325

RESUMO

Among glycosaminoglycan (GAG) biosynthetic enzymes, the human ß1,4-galactosyltransferase 7 (hß4GalT7) is characterized by its unique capacity to take over xyloside derivatives linked to a hydrophobic aglycone as substrates and/or inhibitors. This glycosyltransferase is thus a prime target for the development of regulators of GAG synthesis in therapeutics. Here, we report the structure-guided design of hß4GalT7 inhibitors. By combining molecular modeling, in vitro mutagenesis, and kinetic measurements, and in cellulo analysis of GAG anabolism and decorin glycosylation, we mapped the organization of the acceptor binding pocket, in complex with 4-methylumbelliferone-xylopyranoside as prototype substrate. We show that its organization is governed, on one side, by three tyrosine residues, Tyr(194), Tyr(196), and Tyr(199), which create a hydrophobic environment and provide stacking interactions with both xylopyranoside and aglycone rings. On the opposite side, a hydrogen-bond network is established between the charged amino acids Asp(228), Asp(229), and Arg(226), and the hydroxyl groups of xylose. We identified two key structural features, i.e. the strategic position of Tyr(194) forming stacking interactions with the aglycone, and the hydrogen bond between the His(195) nitrogen backbone and the carbonyl group of the coumarinyl molecule to develop a tight binder of hß4GalT7. This led to the synthesis of 4-deoxy-4-fluoroxylose linked to 4-methylumbelliferone that inhibited hß4GalT7 activity in vitro with a Ki 10 times lower than the Km value and efficiently impaired GAG synthesis in a cell assay. This study provides a valuable probe for the investigation of GAG biology and opens avenues toward the development of bioactive compounds to correct GAG synthesis disorders implicated in different types of malignancies.


Assuntos
Inibidores Enzimáticos/química , Galactosiltransferases/metabolismo , Xilosidases/antagonistas & inibidores , Domínio Catalítico , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Galactosiltransferases/química , Humanos , Cinética , Modelos Moleculares , Sondas Moleculares
3.
Am J Respir Cell Mol Biol ; 50(3): 583-94, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24127863

RESUMO

Glycosaminoglycan (GAG) chains of proteoglycans (PGs) play important roles in fibrosis through cell-matrix interactions and growth factor binding in the extracellular matrix. We investigated the expression and regulation of PG core protein (versican) and key enzymes (xylosyltransferase [XT]-I, ß1,3-glucuronosyltransferase [GlcAT]-I, chondroitin-4-sulfotransferase [C4ST]) implicated in synthesis and sulfation of GAGs in bleomycin (BLM) and adenovirus-transforming growth factor (TGF)-ß1-induced lung fibrosis in rats. We also studied the role of GlcAT-I or TGF-ß1 and the signaling pathways regulating PG-GAG production in primary lung fibroblasts isolated from saline- or BLM-instilled rats. The mRNA for XT-I, GlcAT-I, C4ST, and versican was increased in the lung 14 days after BLM injury. In vitro studies indicate that fibrotic lung fibroblasts (FLFs) expressed more XT-I, C4ST, and chondroitin sulfate (CS)-GAGs than did normal lung fibroblasts at baseline. TGF-ß1 enhanced the expression of XT-I, C4ST-I, and versican in normal lung fibroblasts, whereas SB203580 or SB431542, by targeting p38 mitogen-activated protein kinase or TGF-ß type-1 receptor/activin receptor-like kinase 5, respectively, attenuated the response to both TGF-ß1 and FLFs on PG-GAG expression. Neutralizing anti-TGF-ß1 antibody abrogated FLF-conditioned medium-stimulated expression of XT-I, GlcAT-I, versican, and CS-GAG. Forced expression of TGF-ß1 in vivo enhanced versican, XT-I, GlcAT-I, and C4ST-I expression and PG-GAG deposition in rat lungs. Finally, induced expression of GlcAT-I gene in rat lung fibroblasts increased GAG synthesis by these cells. Together, our results provide new insights into the basis for increased PG-GAG deposition in lung fibrosis; inhibition of TGF-ß1-mediated or fibrosis-induced PG-GAG production by activin receptor-like kinase 5/p38 inhibitors may contribute to antifibrotic activity.


Assuntos
Bleomicina , Glicosaminoglicanos/metabolismo , Glicosiltransferases/metabolismo , Pulmão/enzimologia , Fibrose Pulmonar/enzimologia , Fator de Crescimento Transformador beta1/metabolismo , Animais , Anticorpos Neutralizantes/farmacologia , Células Cultivadas , Sulfatos de Condroitina/metabolismo , Modelos Animais de Doenças , Fibroblastos/enzimologia , Fibroblastos/patologia , Regulação Enzimológica da Expressão Gênica , Glucuronosiltransferase/metabolismo , Glicosaminoglicanos/genética , Glicosiltransferases/genética , Pulmão/efeitos dos fármacos , Pulmão/patologia , Masculino , Pentosiltransferases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Fibrose Pulmonar/patologia , Fibrose Pulmonar/prevenção & controle , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Sulfotransferases/metabolismo , Fatores de Tempo , Transfecção , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator de Crescimento Transformador beta1/genética , Regulação para Cima , Versicanas/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , UDP Xilose-Proteína Xilosiltransferase
4.
J Biol Chem ; 288(3): 1774-84, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23223231

RESUMO

Xylosyltransferase I (XT-I) is an essential enzyme of proteoglycan (PG) biosynthesis pathway catalyzing the initial and rate-limiting step in glycosaminoglycan chain assembly. It plays a critical role in the regulation of PG synthesis in cartilage; however, little is known about underlying mechanism. Here, we provide evidence that, in human primary chondrocytes, IL-1ß regulates XT-I gene expression into an early phase of induction and a late phase of down-regulation. Based on promoter deletions, the region up to -850 bp was defined as a major element of XT-I gene displaying both constitutive and IL-1ß-regulated promoter activity. Point mutation and signaling analyses revealed that IL-1ß-induced promoter activity is achieved through AP-1 response elements and mediated by SAP/JNK and p38 signaling pathways. Transactivation and chromatin immunoprecipitation assays indicated that AP-1 is a potent transactivator of XT-I promoter and that IL-1ß-induced activity is mediated through increased recruitment of AP-1 to the promoter. Finally, we show that Sp3 is a repressor of XT-I promoter and bring evidence that the repressive effect of IL-1ß during the late phase is mediated through Sp3 recruitment to the promoter. This suggests that modulation of Sp3 in cartilage could prevent IL-1ß inhibition of PG synthesis and limit tissue degradation.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Pentosiltransferases/genética , Proteoglicanas/biossíntese , Fator de Transcrição Sp3/genética , Fator de Transcrição AP-1/genética , Idoso , Sequência de Bases , Sítios de Ligação , Cartilagem/citologia , Cartilagem/efeitos dos fármacos , Cartilagem/metabolismo , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Humanos , Interleucina-1beta/farmacologia , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mutação , Pentosiltransferases/metabolismo , Cultura Primária de Células , Regiões Promotoras Genéticas , Ligação Proteica , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição Sp3/metabolismo , Fator de Transcrição AP-1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , UDP Xilose-Proteína Xilosiltransferase
5.
Cell Death Dis ; 14(6): 355, 2023 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-37296099

RESUMO

Genetic mutations in the Xylt1 gene are associated with Desbuquois dysplasia type II syndrome characterized by sever prenatal and postnatal short stature. However, the specific role of XylT-I in the growth plate is not completely understood. Here, we show that XylT-I is expressed and critical for the synthesis of proteoglycans in resting and proliferative but not in hypertrophic chondrocytes in the growth plate. We found that loss of XylT-I induces hypertrophic phenotype-like of chondrocytes associated with reduced interterritorial matrix. Mechanistically, deletion of XylT-I impairs the synthesis of long glycosaminoglycan chains leading to the formation of proteoglycans with shorter glycosaminoglycan chains. Histological and Second Harmonic Generation microscopy analysis revealed that deletion of XylT-I accelerated chondrocyte maturation and prevents chondrocytes columnar organization and arrangement in parallel of collagen fibers in the growth plate, suggesting that XylT-I controls chondrocyte maturation and matrix organization. Intriguingly, loss of XylT-I induced at embryonic stage E18.5 the migration of progenitor cells from the perichondrium next to the groove of Ranvier into the central part of epiphysis of E18.5 embryos. These cells characterized by higher expression of glycosaminoglycans exhibit circular organization then undergo hypertrophy and death creating a circular structure at the secondary ossification center location. Our study revealed an uncovered role of XylT-I in the synthesis of proteoglycans and provides evidence that the structure of glycosaminoglycan chains of proteoglycans controls chondrocyte maturation and matrix organization.


Assuntos
Condrócitos , Proteoglicanas , Humanos , Condrócitos/metabolismo , Proteoglicanas/metabolismo , Lâmina de Crescimento , Glicosaminoglicanos , Colágeno/metabolismo , Hipertrofia/metabolismo , UDP Xilose-Proteína Xilosiltransferase
6.
Glycobiology ; 22(4): 561-71, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22156920

RESUMO

Glycosaminoglycan (GAG) assembly initiates through the formation of a linkage tetrasaccharide region serving as a primer for both chondroitin sulfate (CS) and heparan sulfate (HS) chain polymerization. A possible role for sulfation of the linkage structure and of the constitutive disaccharide unit of CS chains in the regulation of CS-GAG chain synthesis has been suggested. To investigate this, we determined whether sulfate substitution of galactose (Gal) residues of the linkage region or of N-acetylgalactosamine (GalNAc) of the disaccharide unit influences activity and specificity of chondroitin sulfate N-acetylgalactosaminyltransferase-1 (CSGalNAcT-1), a key glycosyltransferase of CS biosynthesis. We synthesized a series of sulfated and unsulfated analogs of the linkage oligosaccharide and of the constitutive unit of CS and tested these molecules as potential acceptor substrates for the recombinant human CSGalNAcT-1. We show here that sulfation at C4 or C6 of the Gal residues markedly influences CSGalNAcT-1 initiation activity and catalytic efficiency. Kinetic analysis indicates that CSGalNAcT-1 exhibited 3.6-, 1.6-, and 2.2-fold higher enzymatic efficiency due to lower K(m) values toward monosulfated trisaccharides substituted at C4 or C6 position of Gal1, and at C6 of Gal2, respectively, compared with the unsulfated oligosaccharide. This highlights the critical influence of Gal substitution on both CSGalNAcT-1 activity and specifity. No GalNAcT activity was detected toward sulfated and unsulfated analogs of the CS constitutive disaccharide (GlcA-ß1,3-GalNAc), indicating that CSGalNAcT-1 was involved in initiation but not in elongation of CS chains. Our results strongly suggest that sulfation of the linkage region acts as a regulatory signal in CS chain initiation.


Assuntos
Sulfatos de Condroitina/química , N-Acetilgalactosaminiltransferases/química , Acetilglucosamina/química , Configuração de Carboidratos , Sequência de Carboidratos , Galactanos/química , Galactose/química , Glicosilação , Células HeLa , Humanos , Cinética , Dados de Sequência Molecular , Oligossacarídeos/química , Proteínas Recombinantes/química , Especificidade por Substrato
7.
Front Cell Dev Biol ; 10: 903953, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35693943

RESUMO

Glycosylation is a ubiquitous and universal cellular process in all domains of life. In eukaryotes, many glycosylation pathways occur simultaneously onto proteins and lipids for generating a complex diversity of glycan structures. In humans, severe genetic diseases called Congenital Disorders of Glycosylation (CDG), resulting from glycosylation defects, demonstrate the functional relevance of these processes. No real cure exists so far, but oral administration of specific monosaccharides to bypass the metabolic defects has been used in few CDG, then constituting the simplest and safest treatments. Oral D-Galactose (Gal) therapy was seen as a promising tailored treatment for specific CDG and peculiarly for TMEM165-CDG patients. TMEM165 deficiency not only affects the N-glycosylation process but all the other Golgi-related glycosylation types, then contributing to the singularity of this defect. Our previous results established a link between TMEM165 deficiency and altered Golgi manganese (Mn2+) homeostasis. Besides the fascinating power of MnCl2 supplementation to rescue N-glycosylation in TMEM165-deficient cells, D-Gal supplementation has also been shown to be promising in suppressing the observed N-glycosylation defects. Its effect on the other Golgi glycosylation types, most especially O-glycosylation and glycosaminoglycan (GAG) synthesis, was however unknown. In the present study, we demonstrate the differential impact of D-Gal or MnCl2 supplementation effects on the Golgi glycosylation defects caused by TMEM165 deficiency. Whereas MnCl2 supplementation unambiguously fully rescues the N- and O-linked as well as GAG glycosylations in TMEM165-deficient cells, D-Gal supplementation only rescues the N-linked glycosylation, without any effects on the other Golgi-related glycosylation types. According to these results, we would recommend the use of MnCl2 for TMEM165-CDG therapy.

8.
J Biol Chem ; 285(48): 37342-58, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-20843813

RESUMO

Glycosaminoglycans (GAGs) play a central role in many pathophysiological events, and exogenous xyloside substrates of ß1,4-galactosyltransferase 7 (ß4GalT7), a major enzyme of GAG biosynthesis, have interesting biomedical applications. To predict functional peptide regions important for substrate binding and activity of human ß4GalT7, we conducted a phylogenetic analysis of the ß1,4-galactosyltransferase family and generated a molecular model using the x-ray structure of Drosophila ß4GalT7-UDP as template. Two evolutionary conserved motifs, (163)DVD(165) and (221)FWGWGREDDE(230), are central in the organization of the enzyme active site. This model was challenged by systematic engineering of point mutations, combined with in vitro and ex vivo functional assays. Investigation of the kinetic properties of purified recombinant wild-type ß4GalT7 and selected mutants identified Trp(224) as a key residue governing both donor and acceptor substrate binding. Our results also suggested the involvement of the canonical carboxylate residue Asp(228) acting as general base in the reaction catalyzed by human ß4GalT7. Importantly, ex vivo functional tests demonstrated that regulation of GAG synthesis is highly responsive to modification of these key active site amino acids. Interestingly, engineering mutants at position 224 allowed us to modify the affinity and to modulate the specificity of human ß4GalT7 toward UDP-sugars and xyloside acceptors. Furthermore, the W224H mutant was able to sustain decorin GAG chain substitution but not GAG synthesis from exogenously added xyloside. Altogether, this study provides novel insight into human ß4GalT7 active site functional domains, allowing manipulation of this enzyme critical for the regulation of GAG synthesis. A better understanding of the mechanism underlying GAG assembly paves the way toward GAG-based therapeutics.


Assuntos
Galactosiltransferases/química , Galactosiltransferases/metabolismo , Glicosaminoglicanos/biossíntese , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Domínio Catalítico , Galactosiltransferases/genética , Humanos , Invertebrados/química , Invertebrados/classificação , Invertebrados/enzimologia , Invertebrados/genética , Modelos Moleculares , Dados de Sequência Molecular , Filogenia , Alinhamento de Sequência , Especificidade por Substrato , Vertebrados/classificação , Vertebrados/genética , Vertebrados/metabolismo
9.
Am J Physiol Lung Cell Mol Physiol ; 300(2): L191-203, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21056957

RESUMO

Pulmonary fibrosis (PF) is characterized by increased deposition of proteoglycans (PGs), in particular core proteins. Glycosaminoglycans (GAGs) are key players in tissue repair and fibrosis, and we investigated whether PF is associated with changes in the expression and structure of GAGs as well as in the expression of ß1,3-glucuronosyltransferase I (GlcAT-I), a rate-limiting enzyme in GAG synthesis. Lung biopsies from idiopathic pulmonary fibrosis (IPF) patients and lung tissue from a rat model of bleomycin (BLM)-induced PF were immunostained for chondroitin sulfated-GAGs and GlcAT-I expression. Alterations in disaccharide composition and sulfation of chondroitin/dermatan sulfate (CS/DS) were evaluated by fluorophore-assisted carbohydrate electrophoresis (FACE) in BLM rats. Lung fibroblasts isolated from control (saline-instilled) or BLM rat lungs were assessed for GAG structure and GlcAT-I expression. Disaccharide analysis showed that 4- and 6-sulfated disaccharides were increased in the lungs and lung fibroblasts obtained from fibrotic rats compared with controls. Fibrotic lung fibroblasts and transforming growth factor-ß(1) (TGF-ß(1))-treated normal lung fibroblasts expressed increased amounts of hyaluronan and 4- and 6-sulfated chondroitin, and neutralizing anti-TGF-ß(1) antibody diminished the same. TGF-ß(1) upregulated GlcAT-I and versican expression in lung fibroblasts, and signaling through TGF-ß type I receptor/p38 MAPK was required for TGF-ß(1)-mediated GlcAT-I and CS-GAG expression in fibroblasts. Our data show for the first time increased expression of CS-GAGs and GlcAT-I in IPF, fibrotic rat lungs, and fibrotic lung fibroblasts. These data suggest that alterations of sulfation isomers of CS/DS and upregulation of GlcAT-I contribute to the pathological PG-GAG accumulation in PF.


Assuntos
Sulfatos de Condroitina/metabolismo , Dermatan Sulfato/metabolismo , Glucuronosiltransferase/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Animais , Bleomicina/toxicidade , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Glucuronosiltransferase/genética , Humanos , Ácido Hialurônico/metabolismo , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , Pulmão/patologia , Masculino , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator de Crescimento Transformador beta1/farmacologia , Regulação para Cima/efeitos dos fármacos
10.
FASEB J ; 24(2): 436-50, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19812376

RESUMO

Heparan sulfate proteoglycans (HSPGs), strategically located at the cell-tissue-organ interface, regulate major biological processes, including cell proliferation, migration, and adhesion. These vital functions are compromised in tumors, due, in part, to alterations in heparan sulfate (HS) expression and structure. How these modifications occur is largely unknown. Here, we investigated whether epigenetic abnormalities involving aberrant DNA methylation affect HS biosynthetic enzymes in cancer cells. Analysis of the methylation status of glycosyltransferase and sulfotransferase genes in H-HEMC-SS chondrosarcoma cells showed a typical hypermethylation profile of 3-OST sulfotransferase genes. Exposure of chondrosarcoma cells to 5-aza-2'-deoxycytidine (5-Aza-dc), a DNA-methyltransferase inhibitor, up-regulated expression of 3-OST1, 3-OST2, and 3-OST3A mRNAs, indicating that aberrant methylation affects transcription of these genes. Furthermore, HS expression was restored on 5-Aza-dc treatment or reintroduction of 3-OST expression, as shown by indirect immunofluorescence microscopy and/or analysis of HS chains by anion-exchange and gel-filtration chromatography. Notably, 5-Aza-dc treatment of HEMC cells or expression of 3-OST3A cDNA reduced their proliferative and invading properties and augmented adhesion of chondrosarcoma cells. These results provide the first evidence for specific epigenetic regulation of 3-OST genes resulting in altered HSPG sulfation and point to a defect of HS-3-O-sulfation as a factor in cancer progression.


Assuntos
Metilação de DNA , Proteoglicanas de Heparan Sulfato/biossíntese , Sulfotransferases/genética , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Condrossarcoma/genética , Ilhas de CpG/fisiologia , Metilação de DNA/efeitos dos fármacos , Decitabina , Repressão Enzimática , Células HL-60 , Células HeLa , Proteoglicanas de Heparan Sulfato/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Invasividade Neoplásica/genética , Sulfotransferases/efeitos dos fármacos , Regulação para Cima
11.
Cell Death Dis ; 13(1): 11, 2021 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-34930890

RESUMO

TMEM165 deficiency leads to skeletal disorder characterized by major skeletal dysplasia and pronounced dwarfism. However, the molecular mechanisms involved have not been fully understood. Here, we uncover that TMEM165 deficiency impairs the synthesis of proteoglycans by producing a blockage in the elongation of chondroitin-and heparan-sulfate glycosaminoglycan chains leading to the synthesis of proteoglycans with shorter glycosaminoglycan chains. We demonstrated that the blockage in elongation of glycosaminoglycan chains is not due to defect in the Golgi elongating enzymes but rather to availability of the co-factor Mn2+. Supplementation of cell with Mn2+ rescue the elongation process, confirming a role of TMEM165 in Mn2+ Golgi homeostasis. Additionally, we showed that TMEM165 deficiency functionally impairs TGFß and BMP signaling pathways in chondrocytes and in fibroblast cells of TMEM165 deficient patients. Finally, we found that loss of TMEM165 impairs chondrogenic differentiation by accelerating the timing of Ihh expression and promoting early chondrocyte maturation and hypertrophy. Collectively, our results indicate that TMEM165 plays an important role in proteoglycan synthesis and underline the critical role of glycosaminoglycan chains structure in the regulation of chondrogenesis. Our data also suggest that Mn2+ supplementation may be a promising therapeutic strategy in the treatment of TMEM165 deficient patients.


Assuntos
Antiporters/deficiência , Antiporters/metabolismo , Proteínas de Transporte de Cátions/deficiência , Proteínas de Transporte de Cátions/metabolismo , Diferenciação Celular/genética , Condrócitos/metabolismo , Condrócitos/patologia , Sulfatos de Condroitina/biossíntese , Nanismo/metabolismo , Proteoglicanas de Heparan Sulfato/biossíntese , Transdução de Sinais/genética , Animais , Antiporters/genética , Estudos de Casos e Controles , Proteínas de Transporte de Cátions/genética , Linhagem Celular Tumoral , Condrogênese/genética , Nanismo/patologia , Fibroblastos/metabolismo , Técnicas de Inativação de Genes/métodos , Glicosilação , Células HEK293 , Humanos , Hipertrofia/metabolismo , Camundongos , Transfecção
12.
Drug Metab Rev ; 42(1): 159-66, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19807219

RESUMO

The main characteristic of uridine diphosphate (UDP)-glucuronosyltransferases is their potency to glucuronidate a large array of structurally unrelated substances with various nucleophilic groups. The activity of these enzymes strongly depends on their tight association to the membrane of the endoplasmic reticulum. In light of recent data, this review is focused on the membrane-assembly process, which is a prerequisite for activity, and on the amino acids that govern substrate recognition and catalysis at the active site. The major implication of the highly variable N-terminal domain of UDP-glucuronosyltransferases in the substrate specificity of these enzymes is highlighted. In the absence of crystal data of the N-terminal domain, multidisciplinary approaches of genetic-/protein-engineering techniques, homology modeling with glycosyltransferases, and quantitative structure-activity relationships allowed us to point out crucial amino acids. On the basis of these results, possible reaction mechanisms for the glucuronidation of xenobiotics, involving histidine and aspartic acid residues, have been built and are discussed.


Assuntos
Retículo Endoplasmático/química , Glucuronosiltransferase/química , Difosfato de Uridina/química , Animais , Ácido Aspártico/química , Domínio Catalítico , Retículo Endoplasmático/metabolismo , Histidina/química , Membranas/química , Membranas/metabolismo , Conformação Proteica , Relação Quantitativa Estrutura-Atividade , Relação Estrutura-Atividade , Especificidade por Substrato , Xenobióticos/metabolismo
13.
FASEB J ; 23(3): 813-22, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19001053

RESUMO

Osteoarthritis and rheumatoid arthritis are characterized by loss of proteoglycans (PGs) and their glycosaminoglycan (GAG) chains that are essential for cartilage function. Here, we investigated the role of glycosyltransferases (GTs) responsible for PG-GAG chain assembly during joint cartilage destruction and repair processes. At various times after antigen-induced arthritis (AIA) and papain-induced cartilage repair in rats, PG synthesis and deposition, expression of GTs, and GAG chain composition were analyzed. Our data showed that expression of the GT xylosyltransferase I (XT-I) gene initiating PG-GAG chain synthesis was significantly reduced in AIA rat cartilage and was associated with a decrease in PG synthesis. Interestingly, interleukin-1beta, the main proinflammatory cytokine incriminated in joint diseases, down-regulated the XT-I gene expression with a concomitant decrease in PG synthesis in rat cartilage explants ex vivo. However, cartilage from papain-injected rat knees showed up-regulation of XT-I gene expression and increased PG synthesis at early stages of cartilage repair, a process associated with up-regulation of TGF-beta1 gene expression and mediated by p38 mitogen-activated protein kinase activation. Consistently, silencing of XT-I expression by intraarticular injection of XT-I shRNA in rat knees prevented cartilage repair by decreasing PG synthesis and content. These findings show that GTs play a key role in the loss of PG-GAGs in joint diseases and identify novel targets for stimulating cartilage repair.


Assuntos
Cartilagem/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Glicosaminoglicanos/biossíntese , Pentosiltransferases/metabolismo , Agrecanas/genética , Agrecanas/metabolismo , Animais , Artrite/induzido quimicamente , Artrite/metabolismo , Artrite/patologia , Cartilagem/efeitos dos fármacos , Cartilagem/patologia , Inativação Gênica , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Cinética , Masculino , Papaína , Pentosiltransferases/genética , Proteoglicanas/biossíntese , Ratos , Ratos Wistar , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , UDP Xilose-Proteína Xilosiltransferase
14.
Chem Commun (Camb) ; 56(73): 10746-10749, 2020 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-32789356

RESUMO

Here we propose a general strategy to label carbohydrates with N-methyl-anthranilic acid at the anomeric position. Through two examples, we demonstrate that the generated glycoprobes are suitable for fluorescence-based binding/competition assays. Our approach is expected to readily generate series of glycoprobes dedicated to screening assays for the discovery of drugs targeting carbohydrate-protein interactions.


Assuntos
Corantes Fluorescentes/química , Glicosídeos/química , ortoaminobenzoatos/química , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/metabolismo , Glicosídeos/síntese química , Glicosídeos/metabolismo , Proteínas Ligantes de Maltose/metabolismo , Ligação Proteica , Espectrometria de Fluorescência , ortoaminobenzoatos/síntese química , ortoaminobenzoatos/metabolismo
15.
Oncotarget ; 10(34): 3166-3182, 2019 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-31139331

RESUMO

Chondrosarcoma is a highly agressive cancer with currently no effective therapies when unresectable or metastasized, thus the outcome remains poor. High-grade chordrosarcomas are resistant to conventional chemotherapy and radiotherapy and surgical resection remains the only treatment for the majority of chondrosarcomas. Constitutive activation of receptor tyrosine kinases has been shown to be important for malignant transformation and tumour proliferation. Here, we investigated the activation status of EGFR in chondrosarcoma tumor biopsies and cell lines. We found that EGFR is activated in grade II and grade III chondrosarcoma tumors but not in grade I tumors, suggesting a role in tumor progression. Interestingly, we showed that EGFR is activated through an autocrine loop and that inhibition of the EGFR by the TKI, tyrphostin AG1478 or EGFR neutralizing antibodies strongly reduced activation of oncogenic ERK1/2 and mTOR/AKT downstream pathways. Importantly, inhibition of EGFR profoundly reduces cell proliferation and migration, inhibits the expression of MMP13 and MMP3 and enhances cell death. Taken together, these data support the blocking of EGFR as new potential treatment for high-grade chondrosarcoma tumors.

16.
Cell Death Differ ; 25(8): 1442-1456, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29352270

RESUMO

Dysregulation of Wnt signaling has been implicated in developmental defects and in the pathogenesis of many diseases such as osteoarthritis; however, the underlying mechanisms are poorly understood. Here, we report that non-canonical Wnt signaling induced loss of chondrocyte phenotype through activation of Fz-6/DVL-2/SYND4/CaMKIIα/B-raf/ERK1/2 cascade. We show that in response to Wnt-3a, Frizzled 6 (Fz-6) triggers the docking of CaMKIIα to syndecan 4 (SYND4) and that of B-raf to DVL-2, leading to the phosphorylation of B-raf by CaMKIIα and activation of extracellular signal-regulated kinase 1 and 2 (ERK1/2) signaling, which leads to chondrocyte de-differentiation. We demonstrate that CaMKIIα associates and phosphorylates B-raf in vitro and in vivo. Our study reveals the mechanism by which non-canonical Wnt activates ERK1/2 signaling that induces loss of chondrocyte phenotype, and demonstrates a direct functional relationship between CaMKIIα and B-raf during chondrocyte de-differentiation. The identification of Fz-6, SYND4, and B-raf as novel physiological regulators of chondrocyte phenotype may provide new potential anti-osteoarthritic targets.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Desdiferenciação Celular , Proteínas Desgrenhadas/metabolismo , Receptores Frizzled/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Sindecana-4/metabolismo , Proteínas Wnt/metabolismo , Células Cultivadas , Condrócitos/citologia , Condrócitos/metabolismo , Proteínas Desgrenhadas/antagonistas & inibidores , Proteínas Desgrenhadas/genética , Receptores Frizzled/antagonistas & inibidores , Receptores Frizzled/genética , Humanos , Sistema de Sinalização das MAP Quinases , Osteoartrite/metabolismo , Osteoartrite/patologia , Fenótipo , Fosforilação , Proteínas Proto-Oncogênicas B-raf/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Sindecana-4/antagonistas & inibidores , Sindecana-4/genética , beta Catenina/antagonistas & inibidores , beta Catenina/genética , beta Catenina/metabolismo
17.
FEBS J ; 274(5): 1256-64, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17263731

RESUMO

The human UDP-glucuronosyltransferase (UGT) isoforms UGT2B4 and UGT2B7 play a major role in the detoxification of bile acids, steroids and phenols. These two isoforms present distinct but overlapping substrate specificity, sharing common substrates such as the bile acid hyodeoxycholic acid (HDCA) and catechol-estrogens. Here, we show that in UGT2B4, substitution of phenylalanine 33 by leucine suppressed the activity towards HDCA, and impaired the glucuronidation of several substrates, including 4-hydroxyestrone and 17-epiestriol. On the other hand, the substrate specificity of the mutant UGT2B4F33Y, in which phenylalanine was replaced by tyrosine, as found at position 33 of UGT2B7, was similar to wild-type UGT2B4. In the case of UGT2B7, replacement of tyrosine 33 by leucine strongly reduced the activity towards all the tested substrates, with the exception of 17-epiestriol. In contrast, mutation of tyrosine 33 by phenylalanine exhibited similar or even somewhat higher activities than wild-type UGT2B7. Hence, the results strongly indicated that the presence of an aromatic residue at position 33 is important for the activity and substrate specificity of both UGT2B4 and UGT2B7.


Assuntos
Aminoácidos Aromáticos/química , Glucuronosiltransferase/química , Glucuronosiltransferase/metabolismo , Isoenzimas/química , Isoenzimas/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Glucuronosiltransferase/genética , Humanos , Isoenzimas/genética , Cinética , Leucina/metabolismo , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fenilalanina/metabolismo , Homologia de Sequência de Aminoácidos , Spodoptera/citologia , Spodoptera/metabolismo , Especificidade por Substrato , Tirosina/metabolismo
18.
FASEB J ; 20(10): 1692-4, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16807373

RESUMO

The importance of heparan- and chondroitin-sulfate proteoglycans in physiological and pathological processes led to the investigation of the regulation of beta1,3-glucuronosyltransferase I (GlcAT-I), responsible for the completion of glycosaminoglycan-protein linkage tetrasaccharide, a key step prior to polymerization of chondroitin- and heparan-sulfate chains. We have cloned and functionally characterized GlcAT-I 5'-flanking regulatory region. Mutation analysis and electrophoretic mobility shift assays demonstrated the importance of Sp1 motif located at -65/-56 position in promoter activity. Furthermore, we found that elevation of intracellular calcium concentration by the calcium ionophore ionomycin stimulated GlcAT-I gene expression as well as glycosaminoglycan chain synthesis in HeLa cells. Bisanthracycline, an anti-Sp1 compound, inhibited GlcAT-I basal promoter activity and suppressed ionomycin induction, suggesting the importance of Sp1 in calcium induction of GlcAT-I gene expression. Nuclear protein extracts from ionomycin-induced cells exhibited an increased DNA binding of Sp1 factor to the consensus sequence at position -65/-56. Signaling pathway analysis and MEK inhibition studies revealed the important role of p42/p44 MAPK in the stimulation of GlcAT-I promoter activity by ionomycin. The present study identifies, for the first time, GlcAT-I as a target of calcium-dependent signaling pathway and evidences the critical role of Sp1 transcription factor in the activation of GlcAT-I expression.


Assuntos
Cálcio/fisiologia , Regulação da Expressão Gênica , Glucuronosiltransferase/genética , Fator de Transcrição Sp1/metabolismo , Sítios de Ligação , Sulfatos de Condroitina/metabolismo , DNA/metabolismo , Células HeLa , Heparitina Sulfato/metabolismo , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , Ligação Proteica , Proteoglicanas/biossíntese , Transdução de Sinais
19.
Protein Sci ; 15(7): 1667-78, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16815917

RESUMO

The beta1,3-glucuronosyltransferases are responsible for the completion of the protein-glycosaminoglycan linkage region of proteoglycans and of the HNK1 epitope of glycoproteins and glycolipids by transferring glucuronic acid from UDP-alpha-D-glucuronic acid (UDP-GlcA) onto a terminal galactose residue. Here, we develop phylogenetic and mutational approaches to identify critical residues involved in UDP-GlcA binding and enzyme activity of the human beta1,3-glucuronosyltransferase I (GlcAT-I), which plays a key role in glycosaminoglycan biosynthesis. Phylogeny analysis identified 119 related beta1,3-glucuronosyltransferase sequences in vertebrates, invertebrates, and plants that contain eight conserved peptide motifs with 15 highly conserved amino acids. Sequence homology and structural information suggest that Y84, D113, R156, R161, and R310 residues belong to the UDP-GlcA binding site. The importance of these residues is assessed by site-directed mutagenesis, UDP affinity and kinetic analyses. Our data show that uridine binding is primarily governed by stacking interactions with the phenyl group of Y84 and also involves interactions with aspartate 113. Furthermore, we found that R156 is critical for enzyme activity but not for UDP binding, whereas R310 appears less important with regard to both activity and UDP interactions. These results clearly discriminate the function of these two active site residues that were predicted to interact with the pyrophosphate group of UDP-GlcA. Finally, mutation of R161 severely compromises GlcAT-I activity, emphasizing the major contribution of this invariant residue. Altogether, this phylogenetic approach sustained by biochemical analyses affords new insight into the organization of the beta1,3-glucuronosyltransferase family and distinguishes the respective importance of conserved residues in UDP-GlcA binding and activity of GlcAT-I.


Assuntos
Aminoácidos/metabolismo , Glucuronosiltransferase/metabolismo , Mutação , Filogenia , Uridina Difosfato Ácido Glucurônico/metabolismo , Animais , Sítios de Ligação/genética , Sequência Conservada , Glucuronosiltransferase/genética , Humanos , Mutagênese Sítio-Dirigida
20.
Bull Acad Natl Med ; 190(7): 1385-97; discussion 1397-8, 1475-7, 2006 Oct.
Artigo em Francês | MEDLINE | ID: mdl-17450675

RESUMO

Arthritis, osteoarthritis and other degenerative diseases characterized by cartilage deterioration are the most prevalent chronic human health disorders. Despite their major socioeconomic impact there is still no satisfactory treatment. Their frequency is increasing with the lengthening of life expectancy, creating a major public health challenge for coming years. It is important to diagnose such diseases at an early stage and to develop new effective therapies. We are attempting to develop new therapeutic approaches in this context, keeping in mind that cartilage is one of the few human tissues which is unable to regenerate. We intend to identify and characterize key proteins involved in the biosynthesis of cartilage matrix components. One innovative strategy consists of gene transfer, triggering overexpression of native or recombinant factors that can stimulate chondrocyte anabolic activity in order to promote cartilage repair The loss of matrix components, and especially glycosaminoglycans (GAG), is the earliest event in cartilage degeneration. We therefore looked at glycosyltransferases, and especially galactose beta1,3-glucuronosyltransferase-I (GlcAT-1), which catalyses one of the first steps in GAG biosynthesis. We found that any variation in GlcAT-I activity in chondrocytes or cartilage explants (overexpression, or repression with antisense RNA) affected the GAG content of cartilage. Interestingly, overexpression of this enzyme completely counteracted the GAG depletion produced by the proinflammatory cytokine interleukin 1-beta. The neosynthesized GAG was qualitatively identical to that present in the original cartilage matrix. These results are encouraging for therapeutic approaches based on gene transfer We also investigated the structure-function relationship of human recombinant GlcAT-I upon expression in the methyltrophic yeast Pichia pastoris. This allowed us to determine the molecular basis of the recognition of the donor and acceptor substrates of the enzyme. This multidisciplinary research, based on genetic and protein engineering, molecular modelling and glycochemistry will lay the groundwork for designing original glycomimetics able to stimulate GAG synthesis.


Assuntos
Artrite/terapia , Doenças das Cartilagens/terapia , Cartilagem Articular , Condrócitos/enzimologia , Glicosaminoglicanos/biossíntese , Glicosaminoglicanos/metabolismo , Glicosiltransferases/metabolismo , Artrite/metabolismo , Materiais Biocompatíveis , Doenças das Cartilagens/metabolismo , Cartilagem Articular/metabolismo , Cartilagem Articular/fisiologia , Condrócitos/metabolismo , Técnicas de Transferência de Genes , Engenharia Genética , Glicosiltransferases/genética , Homeostase , Humanos , Metaloproteinases da Matriz/metabolismo , Engenharia de Proteínas , Proteoglicanas/metabolismo , RNA Antissenso/farmacologia , Regeneração , Pesquisa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA