Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Cell Death Dis ; 14(6): 355, 2023 06 09.
Article in English | MEDLINE | ID: mdl-37296099

ABSTRACT

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.


Subject(s)
Chondrocytes , Proteoglycans , Humans , Chondrocytes/metabolism , Proteoglycans/metabolism , Growth Plate , Glycosaminoglycans , Collagen/metabolism , Hypertrophy/metabolism , UDP Xylose-Protein Xylosyltransferase
2.
Front Cell Dev Biol ; 10: 903953, 2022.
Article in English | MEDLINE | ID: mdl-35693943

ABSTRACT

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.

3.
Cell Death Dis ; 13(1): 11, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34930890

ABSTRACT

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.


Subject(s)
Antiporters/deficiency , Antiporters/metabolism , Cation Transport Proteins/deficiency , Cation Transport Proteins/metabolism , Cell Differentiation/genetics , Chondrocytes/metabolism , Chondrocytes/pathology , Chondroitin Sulfates/biosynthesis , Dwarfism/metabolism , Heparan Sulfate Proteoglycans/biosynthesis , Signal Transduction/genetics , Animals , Antiporters/genetics , Case-Control Studies , Cation Transport Proteins/genetics , Cell Line, Tumor , Chondrogenesis/genetics , Dwarfism/pathology , Fibroblasts/metabolism , Gene Knockout Techniques/methods , Glycosylation , HEK293 Cells , Humans , Hypertrophy/metabolism , Mice , Transfection
4.
Chem Commun (Camb) ; 56(73): 10746-10749, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32789356

ABSTRACT

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.


Subject(s)
Fluorescent Dyes/chemistry , Glycosides/chemistry , ortho-Aminobenzoates/chemistry , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , Glycosides/chemical synthesis , Glycosides/metabolism , Maltose-Binding Proteins/metabolism , Protein Binding , Spectrometry, Fluorescence , ortho-Aminobenzoates/chemical synthesis , ortho-Aminobenzoates/metabolism
5.
Oncotarget ; 10(34): 3166-3182, 2019 May 07.
Article in English | MEDLINE | ID: mdl-31139331

ABSTRACT

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.

6.
FASEB J ; 33(2): 2669-2679, 2019 02.
Article in English | MEDLINE | ID: mdl-30307768

ABSTRACT

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+.


Subject(s)
Gene Expression Regulation/drug effects , Indoles/pharmacology , Manganese/pharmacology , Membrane Proteins/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Thapsigargin/pharmacology , Antiporters , Biological Transport , Calcium/metabolism , Cation Transport Proteins , Congenital Disorders of Glycosylation/drug therapy , Congenital Disorders of Glycosylation/metabolism , Congenital Disorders of Glycosylation/pathology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/pharmacology , Glycosylation , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , HEK293 Cells , Homeostasis , Humans , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomal-Associated Membrane Protein 2/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics
7.
Cell Death Differ ; 25(8): 1442-1456, 2018 08.
Article in English | MEDLINE | ID: mdl-29352270

ABSTRACT

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.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Dedifferentiation , Dishevelled Proteins/metabolism , Frizzled Receptors/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Syndecan-4/metabolism , Wnt Proteins/metabolism , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Dishevelled Proteins/antagonists & inhibitors , Dishevelled Proteins/genetics , Frizzled Receptors/antagonists & inhibitors , Frizzled Receptors/genetics , Humans , MAP Kinase Signaling System , Osteoarthritis/metabolism , Osteoarthritis/pathology , Phenotype , Phosphorylation , Proto-Oncogene Proteins B-raf/genetics , RNA Interference , RNA, Small Interfering/metabolism , Syndecan-4/antagonists & inhibitors , Syndecan-4/genetics , beta Catenin/antagonists & inhibitors , beta Catenin/genetics , beta Catenin/metabolism
8.
PLoS One ; 11(1): e0146499, 2016.
Article in English | MEDLINE | ID: mdl-26751072

ABSTRACT

Stimulation of proteoglycan (PG) synthesis and deposition plays an important role in the pathophysiology of fibrosis and is an early and dominant feature of pulmonary fibrosis. Transforming growth factor-ß1 (TGF-ß1) is a major cytokine associated with fibrosis that induces excessive synthesis of matrix proteins, particularly PGs. Owing to the importance of PGs in matrix assembly and in mediating cytokine and growth factor signaling, a strategy based on the inhibition of PG synthesis may prevent excessive matrix PG deposition and attenuates profibrotic effects of TGF-ß1 in lung fibroblasts. Here, we showed that 4-MU4-deoxy-ß-D-xylopyranoside, a competitive inhibitor of ß4-galactosyltransferase7, inhibited PG synthesis and secretion in a dose-dependent manner by decreasing the level of both chondroitin/dermatan- and heparin-sulfate PG in primary lung fibroblasts. Importantly, 4-MU4-deoxy-xyloside was able to counteract TGF-ß1-induced synthesis of PGs, activation of fibroblast proliferation and fibroblast-myofibroblast differentiation. Mechanistically, 4-MU4-deoxy-xyloside treatment inhibited TGF-ß1-induced activation of canonical Smads2/3 signaling pathway in lung primary fibroblasts. The knockdown of ß4-galactosyltransferase7 mimicked 4-MU4-deoxy-xyloside effects, indicating selective inhibition of ß4-galactosyltransferase7 by this compound. Collectively, this study reveals the anti-fibrotic activity of 4-MU4-deoxy-xyloside and indicates that inhibition of PG synthesis represents a novel strategy for the treatment of lung fibrosis.


Subject(s)
Hymecromone/analogs & derivatives , Lung/physiopathology , Proteoglycans/biosynthesis , Pulmonary Fibrosis/physiopathology , Transforming Growth Factor beta1/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Cytokines/metabolism , Dermatan Sulfate/chemistry , Enzyme Inhibitors/chemistry , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Galactosyltransferases/antagonists & inhibitors , Glycosides/chemistry , Heparin/analogs & derivatives , Heparin/biosynthesis , Humans , Hymecromone/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Lung/cytology , Lung/metabolism , N-Acetyllactosamine Synthase/antagonists & inhibitors , Phenotype , Pulmonary Fibrosis/drug therapy , RNA, Small Interfering/metabolism , Rats , Real-Time Polymerase Chain Reaction , Signal Transduction
9.
J Biol Chem ; 290(12): 7658-70, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25568325

ABSTRACT

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.


Subject(s)
Enzyme Inhibitors/chemistry , Galactosyltransferases/metabolism , Xylosidases/antagonists & inhibitors , Catalytic Domain , Drug Design , Enzyme Inhibitors/pharmacology , Galactosyltransferases/chemistry , Humans , Kinetics , Models, Molecular , Molecular Probes
10.
Front Cell Neurosci ; 8: 349, 2014.
Article in English | MEDLINE | ID: mdl-25389387

ABSTRACT

UDP-glucuronosyltransferases (UGTs) form a multigenic family of membrane-bound enzymes expressed in various tissues, including brain. They catalyze the formation of ß-D-glucuronides from structurally unrelated substances (drugs, other xenobiotics, as well as endogenous compounds) by the linkage of glucuronic acid from the high energy donor, UDP-α-D-glucuronic acid. In brain, UGTs actively participate to the overall protection of the tissue against the intrusion of potentially harmful lipophilic substances that are metabolized as hydrophilic glucuronides. These metabolites are generally inactive, except for important pharmacologically glucuronides such as morphine-6-glucuronide. UGTs are mainly expressed in endothelial cells and astrocytes of the blood brain barrier (BBB). They are also associated to brain interfaces devoid of BBB, such as circumventricular organ, pineal gland, pituitary gland and neuro-olfactory tissues. Beside their key-role as a detoxication barrier, UGTs play a role in the steady-state of endogenous compounds, like steroids or dopamine (DA) that participate to the function of the brain. UGT isoforms of family 1A, 2A, 2B and 3A are expressed in brain tissues to various levels and are known to present distinct but overlapping substrate specificity. The importance of these enzyme species with regard to the formation of toxic, pharmacologically or physiologically relevant glucuronides in the brain will be discussed.

11.
Am J Respir Cell Mol Biol ; 50(3): 583-94, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24127863

ABSTRACT

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.


Subject(s)
Bleomycin , Glycosaminoglycans/metabolism , Glycosyltransferases/metabolism , Lung/enzymology , Pulmonary Fibrosis/enzymology , Transforming Growth Factor beta1/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Cells, Cultured , Chondroitin Sulfates/metabolism , Disease Models, Animal , Fibroblasts/enzymology , Fibroblasts/pathology , Gene Expression Regulation, Enzymologic , Glucuronosyltransferase/metabolism , Glycosaminoglycans/genetics , Glycosyltransferases/genetics , Lung/drug effects , Lung/pathology , Male , Pentosyltransferases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/prevention & control , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Sulfotransferases/metabolism , Time Factors , Transfection , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/genetics , Up-Regulation , Versicans/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism , UDP Xylose-Protein Xylosyltransferase
12.
J Biol Chem ; 288(3): 1774-84, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23223231

ABSTRACT

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.


Subject(s)
Gene Expression Regulation/drug effects , Pentosyltransferases/genetics , Proteoglycans/biosynthesis , Sp3 Transcription Factor/genetics , Transcription Factor AP-1/genetics , Aged , Base Sequence , Binding Sites , Cartilage/cytology , Cartilage/drug effects , Cartilage/metabolism , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Humans , Interleukin-1beta/pharmacology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Middle Aged , Molecular Sequence Data , Mutation , Pentosyltransferases/metabolism , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Signal Transduction/drug effects , Sp3 Transcription Factor/metabolism , Transcription Factor AP-1/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , UDP Xylose-Protein Xylosyltransferase
13.
PLoS One ; 7(3): e34020, 2012.
Article in English | MEDLINE | ID: mdl-22479506

ABSTRACT

Loss of glycosaminoglycan (GAG) chains of proteoglycans (PGs) is an early event of osteoarthritis (OA) resulting in cartilage degradation that has been previously demonstrated in both huma and experimental OA models. However, the mechanism of GAG loss and the role of xylosyltransferase-I (XT-I) that initiates GAG biosynthesis onto PG molecules in the pathogenic process of human OA are unknown. In this study, we have characterized XT-I expression and activity together with GAG synthesis in human OA cartilage obtained from different regions of the same joint, defined as "normal", "late-stage" or adjacent to "late-stage". The results showed that GAG synthesis and content increased in cartilage from areas flanking OA lesions compared to cartilage from macroscopically "normal" unaffected regions, while decreased in "late-stage" OA cartilage lesions. This increase in anabolic state was associated with a marked upregulation of XT-I expression and activity in cartilage "next to lesion" while a decrease in the "late-stage" OA cartilage. Importantly, XT-I inhibition by shRNA or forced-expression with a pCMV-XT-I construct correlated with the modulation of GAG anabolism in human cartilage explants. The observation that XT-I gene expression was down-regulated by IL-1ß and up-regulated by TGF-ß1 indicates that these cytokines may play a role in regulating GAG content in human OA. Noteworthy, expression of IL-1ß receptor (IL-1R1) was down-regulated whereas that of TGF-ß1 was up-regulated in early OA cartilage. Theses observations may account for upregulation of XT-I and sustained GAG synthesis prior to the development of cartilage lesions during the pathogenic process of OA.


Subject(s)
Gene Expression Regulation, Enzymologic , Glycosaminoglycans/metabolism , Osteoarthritis/metabolism , Pentosyltransferases/physiology , Receptors, Interleukin-1 Type I/metabolism , Aged , Cartilage, Articular/metabolism , Disease Progression , Femur/pathology , Gene Expression Profiling , Humans , Interleukin-1beta/metabolism , Middle Aged , Models, Biological , Transforming Growth Factor beta1/metabolism , UDP Xylose-Protein Xylosyltransferase
14.
Methods Mol Biol ; 836: 339-55, 2012.
Article in English | MEDLINE | ID: mdl-22252645

ABSTRACT

Repair of damaged articular cartilage in osteoarthritis (OA) is a clinical challenge. Because cartilage is an avascular and aneural tissue, normal mechanisms of tissue repair through recruitment of cells to the site of tissue destruction are not feasible. Proteoglycan (PG) depletion induced by the proinflammatory cytokine interleukin-1ß, a principal mediator in OA, is a major factor in the onset and progression of joint destruction. Current symptomatic treatments of OA by anti-inflammatory drugs do not alter the progression of the disease. Various therapeutic strategies have been developed to antagonize the effect of proinflammatory cytokines. However, relatively few studies were conducted to stimulate anabolic activity, in an attempt to enhance cartilage repair. To this aim, a nonviral gene transfer strategy of glycosyltransferases responsible for PG synthesis has been developed and tested for its capacity to promote cartilage PG synthesis and deposition. Transfection of chondrocytes or cartilage explants by the expression vector for the glycosyltransferase ß-1,3-glucuronosyltransferase-I (GlcAT-I) enhanced PG synthesis and deposition in the ECM by promoting the synthesis of chondroitin sulfate GAG chains of the cartilage matrix. This indicates that therapy mediated through GT gene delivery may constitute a new strategy for the treatment of OA.


Subject(s)
Cartilage, Articular/metabolism , Osteoarthritis/metabolism , Osteoarthritis/therapy , Proteoglycans/metabolism , Animals , Cartilage, Articular/enzymology , Cartilage, Articular/injuries , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/cytology , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Humans , Male , Osteoarthritis/enzymology , Osteoarthritis/pathology , Proteoglycans/biosynthesis , Rats , Rats, Wistar
15.
Glycobiology ; 22(4): 561-71, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22156920

ABSTRACT

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.


Subject(s)
Chondroitin Sulfates/chemistry , N-Acetylgalactosaminyltransferases/chemistry , Acetylglucosamine/chemistry , Carbohydrate Conformation , Carbohydrate Sequence , Galactans/chemistry , Galactose/chemistry , Glycosylation , HeLa Cells , Humans , Kinetics , Molecular Sequence Data , Oligosaccharides/chemistry , Recombinant Proteins/chemistry , Substrate Specificity
16.
Drug Metab Pharmacokinet ; 26(4): 341-50, 2011.
Article in English | MEDLINE | ID: mdl-21422672

ABSTRACT

Ferulic acid (FA), a member of the hydroxycinnamate family, is an abundant dietary antioxidant that may offer beneficial effects against cancer, cardiovascular disease, diabetes, osteoarthritis and Alzheimer's disease. In this study, evidence for sulfation and glucuronidation of FA was investigated upon incubation with human liver microsomes and cytosol. Two main glucuronides, M1 (ether O-glucuronide) and M2 (ester acylglucuronide), were formed with a similar affinity (apparent K(m) 3.53 and 5.15 mM, respectively). A phenol sulfoconjugate was also formed with a higher affinity (K(m) 0.53 mM). Identification of the UDP-glucuronosyltransferase (UGT) isoforms involved in FA glucuronidation was investigated with 12 human recombinant enzymes. FA was mainly glucuronidated by UGT1A isoforms and by UGT2B7. UGT1A4, 2B4, 2B15 and 2B17 failed to glucuronidate the substance. Examination of the kinetic constants revealed that FA was mainly glucuronidated by UGT1A1 at the two nucleophilic groups. UGT1A3 was able to glucuronidate these two positions with the same, but low, efficiency. UGT1A6 and 1A8 were involved in the formation of the ether glucuronide only, whereas UGT1A7, 1A10 and 2B7 preferentially glucuronidated the carboxyl group. Moreover, octyl gallate, a marker substrate of UGT1A1, competitively inhibited FA glucuronidation mediated by this isoform. Altogether, the results suggest that FA glucuronidation is primarily mediated by UGT1A1.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Coumaric Acids/metabolism , Glucuronosyltransferase/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Coumaric Acids/pharmacokinetics , Coumaric Acids/pharmacology , Cytosol/enzymology , Cytosol/metabolism , Female , Gallic Acid/analogs & derivatives , Gallic Acid/metabolism , Gallic Acid/pharmacology , Glucuronides/metabolism , Glucuronosyltransferase/genetics , Humans , Isoenzymes/metabolism , Male , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism
17.
Am J Physiol Lung Cell Mol Physiol ; 300(2): L191-203, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21056957

ABSTRACT

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.


Subject(s)
Chondroitin Sulfates/metabolism , Dermatan Sulfate/metabolism , Glucuronosyltransferase/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Animals , Bleomycin/toxicity , Cells, Cultured , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Glucuronosyltransferase/genetics , Humans , Hyaluronic Acid/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Lung/pathology , Male , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Tissue Distribution , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/pharmacology , Up-Regulation/drug effects
18.
J Biol Chem ; 285(48): 37342-58, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-20843813

ABSTRACT

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.


Subject(s)
Galactosyltransferases/chemistry , Galactosyltransferases/metabolism , Glycosaminoglycans/biosynthesis , Amino Acid Motifs , Amino Acid Sequence , Animals , Catalytic Domain , Galactosyltransferases/genetics , Humans , Invertebrates/chemistry , Invertebrates/classification , Invertebrates/enzymology , Invertebrates/genetics , Models, Molecular , Molecular Sequence Data , Phylogeny , Sequence Alignment , Substrate Specificity , Vertebrates/classification , Vertebrates/genetics , Vertebrates/metabolism
19.
FEBS Lett ; 584(18): 3962-8, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20691685

ABSTRACT

ß1,4-Galactosyltransferase 7 (ß4GalT7) is a key enzyme initiating glycosaminoglycan (GAG) synthesis. Based on in vitro and ex vivo kinetics studies and structure-based modelling, we molecularly characterized ß4GalT7 mutants linked to the progeroid form of Ehlers-Danlos syndrome (EDS), a severe connective tissue disorder. Our results revealed that loss of activity upon L206P substitution due to altered protein folding is the primary cause for the GAG synthesis defect in patients carrying the compound A186D and L206P mutations. We showed that R270C substitution strongly reduced ß4GalT7 affinity towards xyloside acceptor, thus affecting GAG chains formation. This study establishes the molecular basis for ß4GalT7 defects associated with altered GAG synthesis in EDS.


Subject(s)
Ehlers-Danlos Syndrome/enzymology , Galactosyltransferases/metabolism , Glycosaminoglycans/biosynthesis , Animals , CHO Cells , Cricetinae , Cricetulus , Ehlers-Danlos Syndrome/genetics , Galactosyltransferases/chemistry , Galactosyltransferases/genetics , Humans , Models, Molecular , Mutation
20.
Methods Mol Biol ; 640: 309-26, 2010.
Article in English | MEDLINE | ID: mdl-20645060

ABSTRACT

Since phase II reactions quantitatively represent the most important pathways involved in drug biotransformation, the development and the use of in vitro approaches to predict glucuronidation and sulfation are currently attracting intense interest to assist in the selection of new drug candidates and for the optimization of dosage regimens for established drugs. At present, primary cultures of human hepatocytes represent the most suitable in vitro model for drug metabolism studies. This system theoretically expresses the full complement of drug-metabolizing enzymes associated with the endoplasmic reticulum (CYP and UDP-glucuronosyltransferases) or located in the cytosolic compartment (sulfotransferases), and relevant accessory proteins required for drug transport and excretion. Primary hepatocytes also represent a unique in vitro model for global examination of inductive potential of drugs on conjugation reactions (monitored as increases in mRNA content or activity). The progress in cryopreservation over the last decade has made available preserved hepatocytes to address key issues such as the (i) establishment of phase II metabolic profile and rate, (ii) identification of conjugation enzymes involved, and (iii) evaluation of drug-drug interactions. These advances allow a better assessment of phase II reactions during drug discovery and development.


Subject(s)
Glucuronosyltransferase/metabolism , Hepatocytes/enzymology , Pharmaceutical Preparations/metabolism , Sulfotransferases/metabolism , Cells, Cultured , Drug Interactions , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/drug effects , Liver/enzymology , Liver/metabolism , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...