Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 249
Filtrar
1.
Metab Brain Dis ; 38(6): 2133-2144, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37195412

RESUMEN

Mucopolysaccharidoses (MPS) are a group of lysosomal storage diseases (LSD) caused by mutations in genes coding for enzymes responsible for degradation of glycosaminoglycans (GAGs). Most types of these severe disorders are characterized by neuronopathic phenotypes. Although lysosomal accumulation of GAGs is the primary metabolic defect in MPS, secondary alterations in biochemical processes are considerable and influence the course of the disease. Early hypothesis suggested that these secondary changes might be due to lysosomal storage-mediated impairment of activities of other enzymes, and subsequent accumulation of various compounds in cells. However, recent studies indicated that expression of hundreds of genes is changed in MPS cells. Therefore, we asked whether metabolic effects observed in MPS are caused primarily by GAG-mediated inhibition of specific biochemical reactions or appear as results of dysregulation of expression of genes coding for proteins involved in metabolic processes. Transcriptomic analyses of 11 types of MPS (using RNA isolated from patient-derived fibroblasts), performed in this study, showed that a battery of the above mentioned genes is dysregulated in MPS cells. Some biochemical pathways might be especially affected by changes in expression of many genes, including GAG metabolism and sphingolipid metabolism which is especially interesting as secondary accumulation of various sphingolipids is one of the best known additional (while significantly enhancing neuropathological effects) metabolic defects in MPS. We conclude that severe metabolic disturbances, observed in MPS cells, can partially arise from changes in the expression of many genes coding for proteins involved in metabolic processes.


Asunto(s)
Mucopolisacaridosis , Transcriptoma , Humanos , Transcriptoma/genética , Mucopolisacaridosis/genética , Mucopolisacaridosis/patología , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , Línea Celular , Lisosomas/metabolismo
2.
Gene Ther ; 30(1-2): 107-114, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35581402

RESUMEN

Mucopolysaccharidosis IVA (MPS IVA) is a lysosomal storage disorder (LSD) caused by mutations in gene encoding for GALNS enzyme. Lack of GALNS activity leads to the accumulation of glycosaminoglycans (GAGs) keratan sulfate and chondroitin 6-sulfate. Although enzyme replacement therapy has been approved since 2014 for MPS IVA, still there is an unmet medical need to have improved therapies for this disorder. CRISPR/Cas9-based gene therapy has been tested for several LSDs with encouraging findings, but to date it has not been assayed on MPS IVA. In this work, we validated for the first time the use of CRISPR/Cas9, using a Cas9 nickase, for the knock-in of an expression cassette containing GALNS cDNA in an in vitro model of MPS IVA. The results showed the successful homologous recombination of the expression cassette into the AAVS1 locus, as well as a long-term increase in GALNS activity reaching up to 40% of wild-type levels. We also observed normalization of lysosomal mass, total GAGs, and oxidative stress, which are some of the major findings regarding the pathophysiological events in MPS IVA. These results represent a proof-of-concept of the use of CRISPR/Cas9 nickase strategy for the development of a novel therapeutic alternative for MPS IVA.


Asunto(s)
Condroitinsulfatasas , Mucopolisacaridosis IV , Humanos , Mucopolisacaridosis IV/genética , Mucopolisacaridosis IV/terapia , Sistemas CRISPR-Cas , Edición Génica , Condroitinsulfatasas/genética , Condroitinsulfatasas/metabolismo , Condroitinsulfatasas/uso terapéutico , Sulfato de Queratano/metabolismo , Sulfato de Queratano/uso terapéutico , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo
3.
Int J Mol Sci ; 23(17)2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-36077388

RESUMEN

Mucopolysaccharidosis (MPS) is a lysosomal storage disease caused by genetic defects that result in deficiency of one specific enzyme activity, consequently impairing the stepwise degradation of glycosaminoglycans (GAGs). Except for MPS II, the other types of MPS have autosomal recessive inheritance in which two copies of an abnormal allele must be present in order for the disease to develop. In this study, we present the status of variant alleles and biochemistry results found in infants suspected of having MPS I, II, IVA, and VI. A total of 324 suspected infants, including 12 for MPS I, 223 for MPS II, 72 for MPS IVA, and 17 for MPS VI, who were referred for MPS confirmation from newborn screening centers in Taiwan, were enrolled. In all of these infants, one specific enzyme activity in dried blood spot filter paper was lower than the cut-off value in the first blood sample, as well asin a second follow-up sample. The confirmatory methods used in this study included Sanger sequencing, next-generation sequencing, leukocyte enzyme fluorometric assay, and GAG-derived disaccharides in urine using tandem mass spectrometry assays. The results showed that five, nine, and six infants had MPS I, II, and IVA, respectively, and all of them were asymptomatic. Thus, a laboratory diagnosis is extremely important to confirm the diagnosis of MPS. The other infants with identified nucleotide variations and reductions in leukocyte enzyme activities were categorized as being highly suspected cases requiring long-term and intensive follow-up examinations. In summary, the final confirmation of MPS depends on the most powerful biomarkers found in urine, i.e., the quantification of GAG-derived disaccharides including dermatan sulfate, heparan sulfate, and keratan sulfate, and analysis of genetic variants can help predict outcomes and guide treatment.


Asunto(s)
Mucopolisacaridosis , Mucopolisacaridosis II , Mucopolisacaridosis I , Disacáridos , Glicosaminoglicanos/genética , Humanos , Lactante , Recién Nacido , Mucopolisacaridosis/diagnóstico , Mucopolisacaridosis/genética , Espectrometría de Masas en Tándem/métodos
4.
Genes (Basel) ; 13(4)2022 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-35456399

RESUMEN

Monogenic diseases are primarily caused by mutations in a single gene; thus, they are commonly recognized as genetic disorders with the simplest mechanisms. However, recent studies have indicated that the molecular mechanisms of monogenic diseases can be unexpectedly complicated, and their understanding requires complex studies at the molecular level. Previously, we have demonstrated that in mucopolysaccharidoses (MPS), a group of monogenic lysosomal storage diseases, several hundreds of genes reveal significant changes in the expression of various genes. Although the secondary effects of the primary biochemical defect and the inefficient degradation of glycosaminoglycans (GAGs) might be considered, the scale of the changes in the expression of a large fraction of genes cannot be explained by a block in one biochemical pathway. Here, we demonstrate that in cellular models of 11 types of MPS, the expression of genes coding for proteins involved in the regulation of the expression of many other genes at various stages (such as signal transduction, transcription, splicing, RNA degradation, translation, and others) is significantly disturbed relative to the control cells. This conclusion was based on transcriptomic studies, supported by biochemical analyses of levels of selected proteins encoded by genes revealing an especially high level of dysregulation in MPS (EXOSC9, SRSF10, RPL23, and NOTCH3 proteins were investigated). Interestingly, the reduction in GAGs levels, through the inhibition of their synthesis normalized the amounts of EXOSC9, RPL23, and NOTCH3 in some (but not all) MPS types, while the levels of SRSF10 could not be corrected in this way. These results indicate that different mechanisms are involved in the dysregulation of the expression of various genes in MPS, pointing to a potential explanation for the inability of some therapies (such as enzyme replacement therapy or substrate reduction therapy) to fully correct the physiology of MPS patients. We suggest that the disturbed expression of some genes, which appears as secondary or tertiary effects of GAG storage, might not be reversible, even after a reduction in the amounts of the storage material.


Asunto(s)
Enfermedades por Almacenamiento Lisosomal , Mucopolisacaridosis , Proteínas de Ciclo Celular/genética , Regulación de la Expresión Génica/genética , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , Humanos , Mucopolisacaridosis/genética , Mucopolisacaridosis/metabolismo , Proteínas Represoras/genética , Factores de Empalme Serina-Arginina/genética , Transcriptoma
5.
Hum Genet ; 141(7): 1287-1298, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34999954

RESUMEN

SLC10A7, encoded by the so-called SLC10A7 gene, is the seventh member of a human sodium/bile acid cotransporter family, known as the SLC10 family. Despite similarities with the other members of the SLC10 family, SLC10A7 does not exhibit any transport activity for the typical SLC10 substrates and is then considered yet as an orphan carrier. Recently, SLC10A7 mutations have been identified as responsible for a new Congenital Disorder of Glycosylation (CDG). CDG are a family of rare and inherited metabolic disorders, where glycosylation abnormalities lead to multisystemic defects. SLC10A7-CDG patients presented skeletal dysplasia with multiple large joint dislocations, short stature and amelogenesis imperfecta likely mediated by glycosaminoglycan (GAG) defects. Although it has been demonstrated that the transporter and substrate specificities of SLC10A7, if any, differ from those of the main members of the protein family, SLC10A7 seems to play a role in Ca2+ regulation and is involved in proper glycosaminoglycan biosynthesis, especially heparan-sulfate, and N-glycosylation. This paper will review our current knowledge on the known and predicted structural and functional properties of this fascinating protein, and its link with the glycosylation process.


Asunto(s)
Amelogénesis Imperfecta , Trastornos Congénitos de Glicosilación , Osteocondrodisplasias , Simportadores , Trastornos Congénitos de Glicosilación/genética , Glicosaminoglicanos/genética , Glicosilación , Humanos , Transportadores de Anión Orgánico Sodio-Dependiente
6.
J Med Genet ; 59(10): 957-964, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34916232

RESUMEN

BACKGROUND: Mucopolysaccharidoses (MPS) are monogenic metabolic disorders that significantly affect the skeleton. Eleven enzyme defects in the lysosomal degradation of glycosaminoglycans (GAGs) have been assigned to the known MPS subtypes (I-IX). Arylsulfatase K (ARSK) is a recently characterised lysosomal hydrolase involved in GAG degradation that removes the 2-O-sulfate group from 2-sulfoglucuronate. Knockout of Arsk in mice was consistent with mild storage pathology, but no human phenotype has yet been described. METHODS: In this study, we report four affected individuals of two unrelated consanguineous families with homozygous variants c.250C>T, p.(Arg84Cys) and c.560T>A, p.(Leu187Ter) in ARSK, respectively. Functional consequences of the two ARSK variants were assessed by mutation-specific ARSK constructs derived by site-directed mutagenesis, which were ectopically expressed in HT1080 cells. Urinary GAG excretion was analysed by dimethylene blue and electrophoresis, as well as liquid chromatography/mass spectrometry (LC-MS)/MS analysis. RESULTS: The phenotypes of the affected individuals include MPS features, such as short stature, coarse facial features and dysostosis multiplex. Reverse phenotyping in two of the four individuals revealed additional cardiac and ophthalmological abnormalities. Mild elevation of dermatan sulfate was detected in the two subjects investigated by LC-MS/MS. Human HT1080 cells expressing the ARSK-Leu187Ter construct exhibited absent protein levels by western blot, and cells with the ARSK-Arg84Cys construct showed markedly reduced enzyme activity in an ARSK-specific enzymatic assay against 2-O-sulfoglucuronate-containing disaccharides as analysed by C18-reversed-phase chromatography followed by MS. CONCLUSION: Our work provides a detailed clinical and molecular characterisation of a novel subtype of mucopolysaccharidosis, which we suggest to designate subtype X.


Asunto(s)
Arilsulfatasas , Mucopolisacaridosis , Animales , Cromatografía Liquida/métodos , Dermatán Sulfato , Disacáridos/análisis , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , Humanos , Ratones , Ratones Noqueados , Sulfatos , Espectrometría de Masas en Tándem/métodos
7.
ACS Chem Biol ; 17(1): 159-170, 2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-34931806

RESUMEN

Bio-orthogonal chemistries have revolutionized many fields. For example, metabolic chemical reporters (MCRs) of glycosylation are analogues of monosaccharides that contain a bio-orthogonal functionality, such as azides or alkynes. MCRs are metabolically incorporated into glycoproteins by living systems, and bio-orthogonal reactions can be subsequently employed to install visualization and enrichment tags. Unfortunately, most MCRs are not selective for one class of glycosylation (e.g., N-linked vs O-linked), complicating the types of information that can be gleaned. We and others have successfully created MCRs that are selective for intracellular O-GlcNAc modification by altering the structure of the MCR and thus biasing it to certain metabolic pathways and/or O-GlcNAc transferase (OGT). Here, we attempt to do the same for the core GalNAc residue of mucin O-linked glycosylation. The most widely applied MCR for mucin O-linked glycosylation, GalNAz, can be enzymatically epimerized at the 4-hydroxyl to give GlcNAz. This results in a mixture of cell-surface and O-GlcNAc labeling. We reasoned that replacing the 4-hydroxyl of GalNAz with a fluorine would lock the stereochemistry of this position in place, causing the MCR to be more selective. After synthesis, we found that 4FGalNAz labels a variety of proteins in mammalian cells and does not perturb endogenous glycosylation pathways unlike 4FGalNAc. However, through subsequent proteomic and biochemical characterization, we found that 4FGalNAz does not widely label cell-surface glycoproteins but instead is primarily a substrate for OGT. Although these results are somewhat unexpected, they once again highlight the large substrate flexibility of OGT, with interesting and important implications for intracellular protein modification by a potential range of abiotic and native monosaccharides.


Asunto(s)
Acetilglucosamina/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Acetilglucosamina/genética , Animales , Células CHO , Cricetinae , Cricetulus , Galactoquinasa/genética , Galactoquinasa/metabolismo , Galactosiltransferasas/genética , Galactosiltransferasas/metabolismo , Regulación de la Expresión Génica , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , N-Acetilglucosaminiltransferasas/genética , Proteínas Recombinantes , Especificidad por Sustrato , Azúcares de Uridina Difosfato
8.
Int J Mol Sci ; 22(22)2021 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-34830490

RESUMEN

Myopia is the second leading cause of visual impairment globally. Myopia can induce sight-threatening retinal degeneration and the underlying mechanism remains poorly defined. We generated a model of myopia-induced early-stage retinal degeneration in guinea pigs and investigated the mechanism of action. Methods: The form-deprivation-induced myopia (FDM) was induced in the right eyes of 2~3-week-old guinea pigs using a translucent balloon for 15 weeks. The left eye remained untreated and served as a self-control. Another group of untreated age-matched animals was used as naïve controls. The refractive error and ocular biometrics were measured at 3, 7, 9, 12 and 15 weeks post-FDM induction. Visual function was evaluated by electroretinography. Retinal neurons and synaptic structures were examined by confocal microscopy of immunolabelled retinal sections. The total RNAs were extracted from the retinas and processed for RNA sequencing analysis. Results: The FDM eyes presented a progressive axial length elongation and refractive error development. After 15 weeks of intervention, the average refractive power was -3.40 ± 1.85 D in the FDM eyes, +2.94 ± 0.59 D and +2.69 ± 0.56 D in the self-control and naïve control eyes, respectively. The a-wave amplitude was significantly lower in FDM eyes and these eyes had a significantly lower number of rods, secretagogin+ bipolar cells, and GABAergic amacrine cells in selected retinal areas. RNA-seq analysis showed that 288 genes were upregulated and 119 genes were downregulated in FDM retinas compared to naïve control retinas. In addition, 152 genes were upregulated and 12 were downregulated in FDM retinas compared to self-control retinas. The KEGG enrichment analysis showed that tyrosine metabolism, ABC transporters and inflammatory pathways were upregulated, whereas tight junction, lipid and glycosaminoglycan biosynthesis were downregulated in FDM eyes. Conclusions: The long-term (15-week) FDM in the guinea pig models induced an early-stage retinal degeneration. The dysregulation of the tyrosine metabolism and inflammatory pathways may contribute to the pathogenesis of myopia-induced retinal degeneration.


Asunto(s)
Inflamación/genética , Miopía/genética , Degeneración Retiniana/genética , Tirosina/metabolismo , Animales , Modelos Animales de Enfermedad , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , Cobayas , Humanos , Inflamación/patología , Redes y Vías Metabólicas/genética , Miopía/complicaciones , Miopía/patología , RNA-Seq , Retina/metabolismo , Retina/patología , Degeneración Retiniana/etiología , Degeneración Retiniana/patología , Tirosina/genética
9.
Int J Mol Sci ; 22(22)2021 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-34830113

RESUMEN

Mucopolysaccharidosis type II (MPS II) results from the dysfunction of a lysosomal enzyme, iduronate-2-sulfatase (IDS). Dysfunction of IDS triggers the lysosomal accumulation of its substrates, glycosaminoglycans, leading to mental retardation and systemic symptoms including skeletal deformities and valvular heart disease. Most patients with severe types of MPS II die before the age of 20. The administration of recombinant IDS and transplantation of hematopoietic stem cells are performed as therapies for MPS II. However, these therapies either cannot improve functions of the central nervous system or cause severe side effects, respectively. To date, 729 pathogenetic variants in the IDS gene have been reported. Most of these potentially cause misfolding of the encoded IDS protein. The misfolded IDS mutants accumulate in the endoplasmic reticulum (ER), followed by degradation via ER-associated degradation (ERAD). Inhibition of the ERAD pathway or refolding of IDS mutants by a molecular chaperone enables recovery of the lysosomal localization and enzyme activity of IDS mutants. In this review, we explain the IDS structure and mechanism of activation, and current findings about the mechanism of degradation-dependent loss of function caused by pathogenetic IDS mutation. We also provide a potential therapeutic approach for MPS II based on this loss-of-function mechanism.


Asunto(s)
Degradación Asociada con el Retículo Endoplásmico , Retículo Endoplásmico , Glicoproteínas , Glicosaminoglicanos , Mucopolisacaridosis II , Mutación , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mucopolisacaridosis II/enzimología , Mucopolisacaridosis II/genética
10.
J Biol Chem ; 297(5): 101293, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34634304

RESUMEN

Golgi-resident bisphosphate nucleotidase 2 (BPNT2) is a member of a family of magnesium-dependent, lithium-inhibited phosphatases that share a three-dimensional structural motif that directly coordinates metal binding to effect phosphate hydrolysis. BPNT2 catalyzes the breakdown of 3'-phosphoadenosine-5'-phosphate, a by-product of glycosaminoglycan (GAG) sulfation. KO of BPNT2 in mice leads to skeletal abnormalities because of impaired GAG sulfation, especially chondroitin-4-sulfation, which is critical for proper extracellular matrix development. Mutations in BPNT2 have also been found to underlie a chondrodysplastic disorder in humans. The precise mechanism by which the loss of BPNT2 impairs sulfation remains unclear. Here, we used mouse embryonic fibroblasts (MEFs) to test the hypothesis that the catalytic activity of BPNT2 is required for GAG sulfation in vitro. We show that a catalytic-dead Bpnt2 construct (D108A) does not rescue impairments in intracellular or secreted sulfated GAGs, including decreased chondroitin-4-sulfate, present in Bpnt2-KO MEFs. We also demonstrate that missense mutations in Bpnt2 adjacent to the catalytic site, which are known to cause chondrodysplasia in humans, recapitulate defects in overall GAG sulfation and chondroitin-4-sulfation in MEF cultures. We further show that treatment of MEFs with lithium (a common psychotropic medication) inhibits GAG sulfation and that this effect depends on the presence of BPNT2. Taken together, this work demonstrates that the catalytic activity of an enzyme potently inhibited by lithium can modulate GAG sulfation and therefore extracellular matrix composition, revealing new insights into lithium pharmacology.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Glicosaminoglicanos/metabolismo , Litio/farmacología , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/metabolismo , Animales , Catálisis , Línea Celular , Glicosaminoglicanos/genética , Ratones , Ratones Noqueados , Monoéster Fosfórico Hidrolasas/genética
11.
Sci Rep ; 11(1): 18235, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34521966

RESUMEN

Helicobacter pylori is a causative pathogen of many gastric and extra-gastric diseases. It has infected about half of the global population. There were no genome-wide association studies (GWAS) for H. pylori infection conducted in Chinese population, who carried different and relatively homogenous strain of H. pylori. In this work, we performed SNP (single nucleotide polymorphism)-based, gene-based and pathway-based genome-wide association analyses to investigate the genetic basis of host susceptibility to H. pylori infection in 480 Chinese individuals. We also profiled the composition and function of the gut microbiota between H. pylori infection cases and controls. We found several genes and pathways associated with H. pylori infection (P < 0.05), replicated one previously reported SNP rs10004195 in TLR1 gene region (P = 0.02). We also found that glycosaminoglycan biosynthesis related pathway was associated with both onset and progression of H. pylori infection. In the gut microbiome association study, we identified 2 species, 3 genera and several pathways had differential abundance between H. pylori infected cases and controls. This paper is the first GWAS for H. pylori infection in Chinese population, and we combined the genetic and microbial data to comprehensively discuss the basis of host susceptibility to H. pylori infection.


Asunto(s)
Glicosaminoglicanos/biosíntesis , Infecciones por Helicobacter/genética , Polimorfismo de Nucleótido Simple , Adolescente , Adulto , Femenino , Microbioma Gastrointestinal , Estudio de Asociación del Genoma Completo , Glicosaminoglicanos/genética , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/microbiología , Helicobacter pylori/clasificación , Helicobacter pylori/aislamiento & purificación , Helicobacter pylori/patogenicidad , Humanos , Masculino , Receptor Toll-Like 1/genética
12.
Int J Mol Sci ; 22(6)2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33804258

RESUMEN

The endothelial glycocalyx, the gel layer covering the endothelium, is composed of glycosaminoglycans, proteoglycans, and adsorbed plasma proteins. This structure modulates vessels' mechanotransduction, vascular permeability, and leukocyte adhesion. Thus, it regulates several physiological and pathological events. In the present review, we described the mechanisms that disturb glycocalyx stability such as reactive oxygen species, matrix metalloproteinases, and heparanase. We then focused our attention on the role of glycocalyx degradation in the induction of profibrotic events and on the possible pharmacological strategies to preserve this delicate structure.


Asunto(s)
Endotelio/química , Fibrosis/genética , Glicocálix/química , Mecanotransducción Celular/genética , Proteínas Sanguíneas/química , Proteínas Sanguíneas/genética , Permeabilidad Capilar/genética , Endotelio/ultraestructura , Fibrosis/patología , Glucuronidasa/efectos adversos , Glicocálix/genética , Glicocálix/ultraestructura , Glicosaminoglicanos/química , Glicosaminoglicanos/genética , Humanos , Metaloproteinasas de la Matriz/efectos adversos , Proteoglicanos/química , Proteoglicanos/genética , Especies Reactivas de Oxígeno/efectos adversos
13.
FEBS Lett ; 595(7): 925-941, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33529353

RESUMEN

Pleiotrophin (PTN) is a potent mitogenic cytokine whose activities are controlled by its interactions with glycosaminoglycan (GAG). We examined the specificity of PTN for several types of GAG oligosaccharides. Our data indicate that the interaction of PTN with GAGs is dependent on the sulfation density of GAGs. Surprisingly, an acidic peptide also had similar interactions with PTN as GAGs. This shows that the interaction of PTN with anionic polymers is flexible and adaptable and that the charge density is the main determinant of the interaction. In addition, we show that PTN can compensate for the loss of its termini in interactions with heparin oligosaccharides, allowing it to maintain its affinity for GAGs in the absence of the termini. Taken together, these data provide valuable insight into the interactions of PTN with its proteoglycan receptors.


Asunto(s)
Proteínas Portadoras/genética , Citocinas/genética , Glicosaminoglicanos/genética , Proteoglicanos/genética , Humanos , Oligosacáridos/genética , Péptidos/genética , Unión Proteica/genética
14.
Mol Genet Metab ; 132(3): 180-188, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33558080

RESUMEN

Morquio B disease is an attenuated phenotype within the spectrum of beta galactosidase (GLB1) deficiencies. It is characterised by dysostosis multiplex, ligament laxity, mildly coarse facies and heart valve defects due to keratan sulphate accumulation, predominantly in the cartilage. Morquio B patients have normal neurological development, setting them apart from those with the more severe GM1 gangliosidosis. Morquio B disease, with an incidence of 1:250.000 to 1:1.000.000 live births, is very rare. Here we report the clinical-biochemical data of nine patients. High amounts of keratan sulfate were detected using LC-MS/MS in the patients' urinary samples, while electrophoresis, the standard procedure of qualitative glycosaminoglycans analysis, failed to identify this metabolite in any of the patients' samples. We performed molecular analyses at gene, gene expression and protein expression levels, for both isoforms of the GLB1 gene, lysosomal GLB1, and the cell-surface expressed Elastin Binding Protein. We characterised three novel GLB1 mutations [c.75 + 2 T > G, c.575A > G (p.Tyr192Cys) and c.2030 T > G (p.Val677Gly)] identified in three heterozygous patients. We also set up a copy number variation assay by quantitative PCR to evaluate the presence of deletions/ insertions in the GLB1 gene. We propose a diagnostic plan, setting out the specific clinical- biochemical and molecular features of Morquio B, in order to avoid misdiagnoses and improve patients' management.


Asunto(s)
Gangliosidosis GM1/diagnóstico , Glicosaminoglicanos/genética , Mucopolisacaridosis IV/diagnóstico , beta-Galactosidasa/genética , Niño , Preescolar , Femenino , Gangliosidosis GM1/genética , Gangliosidosis GM1/fisiopatología , Regulación de la Expresión Génica/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Lisosomas/genética , Masculino , Mucopolisacaridosis IV/genética , Mucopolisacaridosis IV/fisiopatología , Mutación Missense/genética , Receptores de Superficie Celular/genética
15.
Sci Rep ; 11(1): 1895, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33479339

RESUMEN

Formation of advanced glycation end products (AGEs), which are associated with diabetes mellitus, contributes to prominent features of osteoarthritis, i.e., inflammation-mediated destruction of articular cartilage. Among the phytochemicals which play a role in anti-inflammatory effects, anthocyanins have also been demonstrated to have anti-diabetic properties. Purple corn is a source of three major anthocyanins: cyanidin-3-O-glucoside, pelargonidin-3-O-glucoside and peonidin-3-O-glucoside. Purple corn anthocyanins have been demonstrated to be involved in the reduction of diabetes-associated inflammation, suggesting that they may have a beneficial effect on diabetes-mediated inflammation of cartilage. This investigation of the chondroprotective effects of purple corn extract on cartilage degradation found a reduction in glycosaminoglycans released from AGEs induced cartilage explants, corresponding with diminishing of uronic acid loss of the cartilage matrix. Investigation of the molecular mechanisms in human articular chondrocytes showed the anti-inflammatory effect of purple corn anthocyanins and the metabolite, protocatechuic acid (PCA) on AGEs induced human articular chondrocytes via inactivation of the NFκb and MAPK signaling pathways. This finding suggests that purple corn anthocyanins and PCA may help ameliorate AGEs mediated inflammation and diabetes-mediated cartilage degradation.


Asunto(s)
Antocianinas/farmacología , Complicaciones de la Diabetes/tratamiento farmacológico , Productos Finales de Glicación Avanzada/genética , Inflamación/tratamiento farmacológico , Antocianinas/química , Cartílago/efectos de los fármacos , Cartílago/patología , Línea Celular , Condrocitos/efectos de los fármacos , Condrocitos/patología , Complicaciones de la Diabetes/genética , Complicaciones de la Diabetes/patología , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Glucósidos/química , Glucósidos/farmacología , Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Glicosaminoglicanos/genética , Humanos , Hidroxibenzoatos/toxicidad , Inflamación/complicaciones , Inflamación/genética , Inflamación/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/genética , Osteoartritis/inducido químicamente , Osteoartritis/tratamiento farmacológico , Osteoartritis/genética , Osteoartritis/patología , Zea mays/química
16.
Mol Biol Rep ; 48(1): 363-370, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33319323

RESUMEN

Mucopolysaccharidosis type I (MPS I) is a lysosomal storage disease caused by a mutation in the IDUA gene, which codes α-L-iduronidase (IDUA), a lysosomal hydrolase that degrades two glycosaminoglycans (GAGs): heparan sulfate (HS) and dermatan sulfate (DS). GAGs are macromolecules found mainly in the extracellular matrix and have important signaling and structural roles which are essential to the maintenance of cell and tissue physiology. Nondegraded GAGs accumulate in various cell types, which characterizes MPS I as a multisystemic progressive disease. Many tissues and vital organs have been described in MPS I models, but there is a lack of studies focused on their effects on the reproductive tract. Our previous studies indicated lower sperm production and morphological damage in the epididymis and accessory glands in male MPS I mice, despite their ability to copulate and to impregnate females. Our aim was to improve the testicular characterization of the MPS I model, with a specific focus on ultrastructural observation of the different cell types that compose the seminiferous tubules and interstitium. We investigated the testicular morphology of 6-month-old male C57BL/6 wild-type (Idua+/+) and MPS I (Idua-/-) mice. We found vacuolated cells widely present in the interstitium and important signs of damage in myoid, Sertoli and Leydig cells. In the cytoplasmic region of Sertoli cells, we found an increased number of vesicles with substrates under digestion and a decreased number of electron-dense vesicles similar to lysosomes, suggesting an impaired flux of substrate degradation. Conclusions: Idua exerts an important role in the morphological maintenance of the seminiferous tubules and the testicular interstitium, which may influence the quality of spermatogenesis, having a greater effect with the progression of the disease.


Asunto(s)
Glicosaminoglicanos/genética , Enfermedades por Almacenamiento Lisosomal/genética , Mucopolisacaridosis I/genética , Células de Sertoli/patología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Células Intersticiales de Cajal/metabolismo , Células Intersticiales de Cajal/patología , Células Intersticiales del Testículo/metabolismo , Células Intersticiales del Testículo/patología , Enfermedades por Almacenamiento Lisosomal/metabolismo , Enfermedades por Almacenamiento Lisosomal/patología , Masculino , Ratones , Mucopolisacaridosis I/metabolismo , Mucopolisacaridosis I/patología , Mutación/genética , Células de Sertoli/metabolismo , Espermatozoides/metabolismo , Espermatozoides/patología
17.
J Histochem Cytochem ; 69(2): 137-155, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32936035

RESUMEN

Advances in reagents, methodologies, analytic platforms, and tools have resulted in a dramatic transformation of the research pathology laboratory. These advances have increased our ability to efficiently generate substantial volumes of data on the expression and accumulation of mRNA, proteins, carbohydrates, signaling pathways, cells, and structures in healthy and diseased tissues that are objective, quantitative, reproducible, and suitable for statistical analysis. The goal of this review is to identify and present how to acquire the critical information required to measure changes in tissues. Included is a brief overview of two morphometric techniques, image analysis and stereology, and the use of artificial intelligence to classify cells and identify hidden patterns and relationships in digital images. In addition, we explore the importance of preanalytical factors in generating high-quality data. This review focuses on techniques we have used to measure proteoglycans, glycosaminoglycans, and immune cells in tissues using immunohistochemistry and in situ hybridization to demonstrate the various morphometric techniques. When performed correctly, quantitative digital pathology is a powerful tool that provides unbiased quantitative data that are difficult to obtain with other methods.


Asunto(s)
Inteligencia Artificial , Glicosaminoglicanos/análisis , Procesamiento de Imagen Asistido por Computador , Proteoglicanos/análisis , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Proteoglicanos/genética , Proteoglicanos/metabolismo
18.
Cells ; 9(12)2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33317052

RESUMEN

Proper processing of collagens COL1 and COL6 is required for normal function of adipose tissue and skeletal muscle. Proteoglycan decorin (DCN) regulates collagen fiber formation. The amino-terminus of DCN is modified with an O-linked glycosaminoglycan (GAG), the function of which has remained unclear. Previously, non-glycanated DCN (ngDCN) was identified as a marker of adipose stromal cells. Here, we identify MMP14 as the metalloprotease that cleaves DCN to generate ngDCN. We demonstrate that mice ubiquitously lacking DCN GAG (ngDCN mice) have reduced matrix rigidity, enlarged adipocytes, fragile skin, as well as skeletal muscle hypotrophy, fibrosis, and dysfunction. Our results indicate that DCN deglycanation results in reduced intracellular DCN-collagen binding and increased production of truncated COL6 chains, leading to aberrant procollagen processing and extracellular localization. This study reveals that the GAG of DCN functions to regulate collagen assembly in adipose tissue and skeletal muscle and uncovers a new mechanism of matrix dysfunction in obesity and aging.


Asunto(s)
Colágeno Tipo I/metabolismo , Colágeno Tipo VI/metabolismo , Decorina/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Células Cultivadas , Colágeno Tipo I/química , Cadena alfa 1 del Colágeno Tipo I , Colágeno Tipo VI/química , Decorina/genética , Matriz Extracelular/metabolismo , Femenino , Glicosaminoglicanos/química , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , Masculino , Ratones , Músculo Esquelético/metabolismo , Piel/patología
19.
Cells ; 9(12)2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33287330

RESUMEN

Background: Mucopolysaccharidosis type I-Hurler (MPS1-H) is a severe genetic lysosomal storage disorder due to loss-of-function mutations in the IDUA gene. The subsequent complete deficiency of alpha l-iduronidase enzyme is directly responsible of a progressive accumulation of glycosaminoglycans (GAG) in lysosomes which affects the functions of many tissues. Consequently, MPS1 is characterized by systemic symptoms (multiorgan dysfunction) including respiratory and cardiac dysfunctions, skeletal abnormalities and early fatal neurodegeneration. Methods: To understand mechanisms underlying MPS1 neuropathology, we generated induced pluripotent stem cells (iPSC) from a MPS1-H patient with loss-of-function mutations in both IDUA alleles. To avoid variability due to different genetic background of iPSC, we established an isogenic control iPSC line by rescuing IDUA expression by a lentivectoral approach. Results: Marked differences between MPS1-H and IDUA-corrected isogenic controls were observed upon neural differentiation. A scratch assay revealed a strong migration defect of MPS1-H cells. Also, there was a massive impact of IDUA deficiency on gene expression (340 genes with an FDR <0.05). Conclusions: Our results demonstrate a hitherto unknown connection between lysosomal degradation, gene expression and neural motility, which might account at least in part for the phenotype of MPS1-H patients.


Asunto(s)
Movimiento Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Mucopolisacaridosis I/metabolismo , Neuronas/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Expresión Génica/genética , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , Humanos , Iduronidasa/genética , Iduronidasa/metabolismo , Lisosomas/genética , Lisosomas/metabolismo , Mucopolisacaridosis I/genética , Mutación/genética , Fenotipo
20.
Matrix Biol ; 94: 95-109, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33002580

RESUMEN

Recent in vitro evidence shows that glycosaminoglycans (GAGs) and proteoglycans (PGs) in bone matrix may functionally be involved in the tissue-level toughness of bone. In this study, we showed the effect of biglycan (Bgn), a small leucine-rich proteoglycan enriched in extracellular matrix of bone and the associated GAG subtype, chondroitin sulfate (CS), on the toughness of bone in vivo, using wild-type (WT) and Bgn deficient mice. The amount of total GAGs and CS in the mineralized compartment of Bgn KO mouse bone matrix decreased significantly, associated with the reduction of the toughness of bone, in comparison with those of WT mice. However, such differences between WT and Bgn KO mice diminished once the bound water was removed from bone matrix. In addition, CS was identified as the major subtype in bone matrix. We then supplemented CS to both WT and Bgn KO mice to test whether supplemental GAGs could improve the tissue-level toughness of bone. After intradermal administration of CS, the toughness of WT bone was greatly improved, with the GAGs and bound water amount in the bone matrix increased, while such improvement was not observed in Bgn KO mice or with supplementation of dermatan sulfate (DS). Moreover, CS supplemented WT mice exhibited higher bone mineral density and reduced osteoclastogenesis. Interestingly, Bgn KO bone did not show such differences irrespective of the intradermal administration of CS. In summary, the results of this study suggest that Bgn and CS in bone matrix play a pivotal role in imparting the toughness to bone most likely via retaining bound water in bone matrix. Moreover, supplementation of CS improves the toughness of bone in mouse models.


Asunto(s)
Biglicano/genética , Matriz Ósea/crecimiento & desarrollo , Glicosaminoglicanos/metabolismo , Proteoglicanos/metabolismo , Animales , Densidad Ósea/efectos de los fármacos , Matriz Ósea/efectos de los fármacos , Matriz Ósea/metabolismo , Huesos/efectos de los fármacos , Huesos/metabolismo , Sulfatos de Condroitina/farmacología , Dermatán Sulfato/farmacología , Matriz Extracelular/genética , Glicosaminoglicanos/genética , Humanos , Ratones , Ratones Noqueados , Proteoglicanos/genética , Agua
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...