Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Sci Rep ; 10(1): 8641, 2020 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-32433574

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Sci Rep ; 10(1): 166, 2020 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-31932597

RESUMEN

The spiny mouse, Acomys cahirinus displays a unique wound healing ability with regeneration of all skin components in a scar-free manner. To identify orchestrators of this regenerative response we have performed proteomic analyses of skin from Acomys and Mus musculus before and after wounding. Of the ~2000 proteins identified many are expressed at similar levels in Acomys and Mus, but there are significant differences. Following wounding in Mus the complement and coagulation cascades, PPAR signaling pathway and ECM-receptor interactions predominate. In Acomys, other pathways predominate including the Wnt, MAPK, the ribosome, proteasome, endocytosis and tight junction pathways. Notable among Acomys specific proteins are several ubiquitin-associated enzymes and kinases, whereas in Mus immuno-modulation proteins characteristic of inflammatory response are unique or more prominent. ECM proteins such as collagens are more highly expressed in Mus, but likely more important is the higher expression of matrix remodeling proteases in Acomys. Another distinctive difference between Acomys and Mus lies in the macrophage-produced arginase 1 is found in Mus whereas arginase 2 is found in Acomys. Thus, we have identified several avenues for experimental approaches whose aim is to reduce the fibrotic response that the typical mammal displays in response to wounding.


Asunto(s)
Cicatriz/metabolismo , Proteoma/análisis , Regeneración , Piel/metabolismo , Cicatrización de Heridas , Animales , Cicatriz/patología , Ratones , Murinae
3.
Mol Ther Methods Clin Dev ; 15: 305-319, 2019 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-31890730

RESUMEN

rAAVrh74.MCK.GALGT2 is a surrogate gene therapy that inhibits muscular dystrophy in multiple animal models. Here, we report on a dose-response study of functional muscle GALGT2 expression as well as toxicity and biodistribution studies after systemic intravenous (i.v.) delivery of rAAVrh74.MCK.GALGT2. A dose of 4.3 × 1014vg/kg (measured with linear DNA standard) resulted in GALGT2-induced glycosylation in the majority of skeletal myofibers throughout the body and in almost all cardiomyocytes, while several lower doses also showed significant muscle glycosylation. No adverse clinical signs or treatment-dependent changes in tissue or organ pathology were noted at 1 or 3 months post-treatment. Blood cell and serum enzyme chemistry measures in treated mice were all within the normal range except for alkaline phosphatase (ALP) activity, which was elevated in serum but not in tissues. Some anti-rAAVrh74 capsid T cell responses were noted at 4 weeks post-treatment, but all such responses were not present at 12 weeks. Using intramuscular delivery, GALGT2-induced muscle glycosylation was increased in Cmah-deficient mice, which have a humanized sialoglycome, relative to wild-type mice, suggesting that use of mice may underestimate GALGT2 activity in human muscle. These data demonstrate safety and high transduction of muscles throughout the body plan with i.v. delivery of rAAVrh74.MCK.GALGT2.

4.
IEEE Trans Nanobioscience ; 15(1): 11-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26829799

RESUMEN

Carbon nanomaterials have become increasingly popular microelectrode materials for neuroscience applications. Here we study how the scale of carbon nanotubes and carbon nanofibers affect neural viability, outgrowth, and adhesion. Carbon nanotubes were deposited on glass coverslips via a layer-by-layer method with polyethylenimine (PEI). Carbonized nanofibers were fabricated by electrospinning SU-8 and pyrolyzing the nanofiber depositions. Additional substrates tested were carbonized and SU-8 thin films and SU-8 nanofibers. Surfaces were O2-plasma treated, coated with varying concentrations of PEI, seeded with E18 rat cortical cells, and examined at 3, 4, and 7 days in vitro (DIV). Neural adhesion was examined at 4 DIV utilizing a parallel plate flow chamber. At 3 DIV, neural viability was lower on the nanofiber and thin film depositions treated with higher PEI concentrations which corresponded with significantly higher zeta potentials (surface charge); this significance was drastically higher on the nanofibers suggesting that the nanostructure may collect more PEI molecules, causing increased toxicity. At 7 DIV, significantly higher neurite outgrowth was observed on SU-8 nanofiber substrates with nanofibers a significant fraction of a neuron's size. No differences were detected for carbonized nanofibers or carbon nanotubes. Both carbonized and SU-8 nanofibers had significantly higher cellular adhesion post-flow in comparison to controls whereas the carbon nanotubes were statistically similar to control substrates. These data suggest a neural cell preference for larger-scale nanomaterials with specific surface treatments. These characteristics could be taken advantage of in the future design and fabrication of neural microelectrodes.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Nanofibras/toxicidad , Nanotubos de Carbono/toxicidad , Neuritas/efectos de los fármacos , Animales , Línea Celular , Nanofibras/química , Nanomedicina , Nanotubos de Carbono/química , Neuritas/fisiología , Ratas
5.
Neurobiol Dis ; 62: 31-43, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24021662

RESUMEN

Recent clinical and pre-clinical studies suggest that both active and passive immunization strategies targeting Aß amyloid may have clinical benefit in Alzheimer's disease. Here, we demonstrate that vaccination of APPswePSEN1dE9 mice with SDPM1, an engineered non-native Aß amyloid-specific binding peptide, lowers brain Aß amyloid plaque burden and brain Aß1-40 and Aß1-42 peptide levels, improves cognitive learning and memory in Morris water maze tests and increases the expression of synaptic brain proteins. This was the case in young mice immunized prior to development of significant brain amyloid burden, and in older mice, where brain amyloid was already present. Active immunization was optimized using ALUM as an adjuvant to stimulate production of anti-SDPM1 and anti-Aß amyloid antibodies. Intracerebral injection of P4D6, an SDPM1 peptide-mimotope antibody, also lowered brain amyloid plaque burden in APPswePSEN1dE9 mice. Additionally, P4D6 inhibited Aß amyloid-mediated toxicity in cultured neuronal cells. The protein sequence of the variable domain within the P4D6 heavy chain was found to mimic a multimer of the SDPM1 peptide motif. These data demonstrate the efficacy of active and passive vaccine strategies to target Aß amyloid oligomers using an engineered peptide-mimotope strategy.


Asunto(s)
Enfermedad de Alzheimer/terapia , Vacunas contra el Alzheimer/uso terapéutico , Péptidos/uso terapéutico , Hidróxido de Aluminio/inmunología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/metabolismo , Animales , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Hipocampo/patología , Inmunización Pasiva , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/metabolismo , Placa Amiloide/patología , Sinapsis/metabolismo , Resultado del Tratamiento , Vacunación
6.
PLoS One ; 8(8): e73224, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23951345

RESUMEN

The dystroglycan complex contains the transmembrane protein ß-dystroglycan and its interacting extracellular mucin-like protein α-dystroglycan. In skeletal muscle fibers, the dystroglycan complex plays an important structural role by linking the cytoskeletal protein dystrophin to laminin in the extracellular matrix. Mutations that affect any of the proteins involved in this structural axis lead to myofiber degeneration and are associated with muscular dystrophies and congenital myopathies. Because loss of dystrophin in Duchenne muscular dystrophy (DMD) leads to an almost complete loss of dystroglycan complexes at the myofiber membrane, it is generally assumed that the vast majority of dystroglycan complexes within skeletal muscle fibers interact with dystrophin. The residual dystroglycan present in dystrophin-deficient muscle is thought to be preserved by utrophin, a structural homolog of dystrophin that is up-regulated in dystrophic muscles. However, we found that dystroglycan complexes are still present at the myofiber membrane in the absence of both dystrophin and utrophin. Our data show that only a minority of dystroglycan complexes associate with dystrophin in wild type muscle. Furthermore, we provide evidence for at least three separate pools of dystroglycan complexes within myofibers that differ in composition and are differentially affected by loss of dystrophin. Our findings indicate a more complex role of dystroglycan in muscle than currently recognized and may help explain differences in disease pathology and severity among myopathies linked to mutations in DAPC members.


Asunto(s)
Distroglicanos/metabolismo , Distrofina/metabolismo , Músculo Esquelético/metabolismo , Mapas de Interacción de Proteínas , Animales , Distroglicanos/análisis , Distrofina/análisis , Distrofina/genética , Eliminación de Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/química , Sinapsis/química , Sinapsis/metabolismo , Utrofina/análisis , Utrofina/genética , Utrofina/metabolismo
7.
PLoS Curr ; 52013 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-23390591

RESUMEN

α dystroglycan (αDG) is part of the dystrophin-associated glycoprotein (DAG) complex, a series of cytoskeletal, transmembrane, and membrane-associated proteins that serve to link the extracellular matrix (ECM) surrounding individual skeletal myofibers to the intracellular F-actin cytoskeleton. Glycosylation and ECM protein binding to αDG are regulated by a number of genes that, when defective, give rise to congenital or limb-girdle forms of muscular dystrophy termed dystroglycanopathies. One such dystroglycanopathy gene is LARGE. Here, we describe a method to produce and purify full-length, furin-resistant, recombinant αDG from CHO cells and CHO cells overexpressing LARGE (CHO-LARGE). In addition, we analyze the O- and N-linked monosaccharide composition of such proteins. αDG purified from CHO-LARGE cells had increased molar content of xylose and fucose relative to CHO, while no significant changes were found in N-linked monosaccharides. Glucuronic acid could not be quantified by the methods used. These studies describe a method to produce and purify the milligram amounts of αDG needed for certain biochemical methods, including monosaccharide analysis. Key words: Dystroglycan, muscular dystrophy, xylose, fucose, laminin, LARGE Correspondence: Paul.Martin@nationwidechildrens.org.

8.
J Proteome Res ; 11(9): 4413-24, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22775139

RESUMEN

Dystroglycan is a major cell surface glycoprotein receptor for the extracellular matrix in skeletal muscle. Defects in dystroglycan glycosylation cause muscular dystrophy and alterations in dystroglycan glycosylation can impact extracellular matrix binding. Here we describe an immunoprecipitation technique that allows isolation of beta dystroglycan with members of the dystrophin-associated protein complex (DAPC) from detergent-solubilized skeletal muscle. Immunoprecipitation, coupled with shotgun proteomics, has allowed us to identify new dystroglycan-associated proteins and define changed associations that occur within the DAPC in dystrophic skeletal muscles. In addition, we describe changes that result from overexpression of Galgt2, a normally synaptic muscle glycosyltransferase that can modify alpha dystroglycan and inhibit the development of muscular dystrophy when it is overexpressed. These studies identify new dystroglycan-associated proteins that may participate in dystroglycan's roles, both positive and negative, in muscular dystrophy.


Asunto(s)
Distroglicanos/metabolismo , Distrofina/genética , Glicosiltransferasas/genética , Músculo Esquelético/química , Proteoma/análisis , Animales , Western Blotting , Distroglicanos/química , Distrofina/metabolismo , Complejo de Proteínas Asociado a la Distrofina/química , Complejo de Proteínas Asociado a la Distrofina/metabolismo , Glicosiltransferasas/metabolismo , Inmunoprecipitación , Interleucina-15/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Proteínas de Microfilamentos/metabolismo , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Mapeo de Interacción de Proteínas , Proteoma/química , Proteoma/metabolismo , Proteómica/métodos , Reproducibilidad de los Resultados
9.
Sci Transl Med ; 2(42): 42ra54, 2010 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-20668298

RESUMEN

During the evolution of humans, an inactivating deletion was introduced in the CMAH (cytidine monophosphate-sialic acid hydroxylase) gene, which eliminated biosynthesis of the common mammalian sialic acid N-glycolylneuraminic acid from all human cells. We found that this human-specific change in sialylation capacity contributes to the marked discrepancy in phenotype between the mdx mouse model for Duchenne muscular dystrophy (DMD) and the human disease. When compared to human patients with DMD, mdx mice show reduced severity or slower development of clinically relevant disease phenotypes, despite lacking dystrophin protein in almost all muscle cells. This is especially true for the loss of ambulation, cardiac and respiratory muscle weakness, and decreased life span, all of which are major phenotypes contributing to DMD morbidity and mortality. These phenotypes occur at an earlier age or to a greater degree in mdx mice that also carry a human-like mutation in the mouse Cmah gene, possibly as a result of reduced strength and expression of the dystrophin-associated glycoprotein complex and increased activation of complement. Cmah-deficient mdx mice are a small-animal model for DMD that better approximates the human glycome and its contributions to muscular dystrophy.


Asunto(s)
Oxigenasas de Función Mixta/genética , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Animales , Humanos , Ratones , Ratones Endogámicos mdx , Ratones Mutantes , Índice de Severidad de la Enfermedad
10.
Arch Biochem Biophys ; 501(2): 221-31, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20599673

RESUMEN

Defects in glycosylation of decorin can result in systemic hereditary disease. A mutation in the galactosyl transferase I gene is the underlying defect of a progeroid form of Ehlers-Danlos syndrome. We have previously described pathological changes in equine systemic proteoglycan accumulation (ESPA, formerly degenerative suspensory ligament desmitis) as consisting of excessive presence of decorin and other proteoglycans in organs and structures with a high content of connective tissue. Using liquid chromatography/mass spectrometry, and one- and two-dimensional immunoblotting we have determined that decorin from ESPA-tendons had a higher molecular weight than decorin from non-affected control tendons. Glycosaminoglycan structure and monosaccharide composition were determined with HPLC analysis of chondroitinase ABC-digested glycosaminoglycans and gas chromatography/mass spectrometry. This analysis revealed an increase in the total content of sulfated disaccharides, particularly due to enhanced sulfation at 6-position of N-acetyl galactosamine (GalNAc) with a subsequent decrease in the ratio of 4-sulfation to 6-sulfation disaccharides in the ESPA decorin. The ESPA-affected decorin also exhibited altered biological activity resulting in (1) diminished binding of TGFbeta1 (and of anti-decorin antibody) to ESPA decorin, and (2) increased expression of TGFbeta1 in ESPA tissues.


Asunto(s)
Síndrome de Ehlers-Danlos/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Enfermedades de los Caballos/metabolismo , Progeria/metabolismo , Proteoglicanos/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN/genética , Decorina , Modelos Animales de Enfermedad , Síndrome de Ehlers-Danlos/genética , Síndrome de Ehlers-Danlos/patología , Proteínas de la Matriz Extracelular/química , Femenino , Glicosilación , Enfermedades de los Caballos/genética , Enfermedades de los Caballos/patología , Caballos , Humanos , Masculino , Polisacáridos/química , Progeria/genética , Progeria/patología , Proteoglicanos/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tendones/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
11.
Mol Cell Neurosci ; 41(4): 448-63, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19442736

RESUMEN

The CT carbohydrate, Neu5Ac/Neu5Gcalpha2,3[GalNAcbeta1,4]Galbeta1,4GlcNAcbeta-, is specifically expressed at the neuromuscular junction in skeletal myofibers of adult vertebrates. When Galgt2, the glycosyltransferase that creates the synaptic beta1,4GalNAc portion of this glycan, is overexpressed in extrasynaptic regions of the myofiber membrane, alpha dystroglycan becomes glycosylated with the CT carbohydrate and this coincides with the ectopic expression of synaptic dystroglycan-binding proteins, including laminin alpha4, laminin alpha5, and utrophin. Here we show that both synaptic and extrasynaptic forms of laminin and agrin have increased binding to the CT carbohydrate compared to sialyl-N-acetyllactosamine, its extrasynaptically expressed precursor. Muscle laminins also show increased binding to CT-glycosylated muscle alpha dystroglycan relative to its non-CT-containing glycoforms. Overexpression of Galgt2 in transgenic mouse skeletal muscle increased the mRNA expression of extracellular matrix (ECM) genes, including agrin and laminin alpha5, as well as utrophin, integrin alpha7, and neuregulin. Increased expression of ECM proteins in Galgt2 transgenic skeletal muscles was partially dependent on utrophin, but utrophin was not required for Galgt2-induced changes in muscle growth or neuromuscular development. These experiments demonstrate that overexpression of a synaptic carbohydrate can increase both ECM binding to alpha dystroglycan and ECM expression in skeletal muscle, and they suggest a mechanism by which Galgt2 overexpression may inhibit muscular dystrophy and affect neuromuscular development.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Glicosiltransferasas/metabolismo , Músculo Esquelético/metabolismo , Unión Neuromuscular/metabolismo , Utrofina/metabolismo , Agrina/metabolismo , Animales , Línea Celular Transformada , Distroglicanos/metabolismo , Distrofina/metabolismo , Regulación de la Expresión Génica/fisiología , Glicosiltransferasas/genética , Humanos , Técnicas In Vitro , Laminas/metabolismo , Ratones , Ratones Transgénicos , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Transfección/métodos , Utrofina/genética
12.
Am J Pathol ; 171(1): 181-99, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17591965

RESUMEN

A number of recent studies have demonstrated therapeutic effects of transgenes on the development of muscle pathology in the mdx mouse model for Duchenne muscular dystrophy, but none have been shown also to be effective in mouse models for laminin alpha2-deficient congenital muscular dystrophy (MDC1A). Here, we show that overexpression of the cytotoxic T cell (CT) GalNAc transferase (Galgt2) is effective in inhibiting the development of muscle pathology in the dy(W) mouse model of MDC1A, much as we had previously shown in mdx animals. Embryonic overexpression of Galgt2 in skeletal muscles using transgenic mice or postnatal overexpression using adeno-associated virus both reduced the extent of muscle pathology in dy(W)/dy(W) skeletal muscle. As with mdx mice, embryonic overexpression of the Galgt2 transgene in dy(W)/dy(W) myofibers inhibited muscle growth, whereas postnatal overexpression did not. Both embryonic and postnatal overexpression of Galgt2 in dy(W)/dy(W) muscle increased the expression of agrin, a protein that, in recombinant form, has been shown to ameliorate disease, whereas laminin alpha1, another disease modifier, was not expressed. Galgt2 over-expression also stimulated the glycosylation of a gly-colipid with the CT carbohydrate, and glycolipids accounted for most of the CT-reactive material in postnatal overexpression experiments. These experiments demonstrate that Galgt2 overexpression is effective in altering disease progression in skeletal muscles of dy(W) mice and should be considered as a therapeutic target in MDC1A.


Asunto(s)
Glicosiltransferasas/metabolismo , Distrofias Musculares/genética , Distrofia Muscular Animal/metabolismo , Agrina/metabolismo , Animales , Modelos Animales de Enfermedad , Glucolípidos/metabolismo , Glicosilación , Laminina/metabolismo , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Músculo Esquelético/metabolismo , Distrofias Musculares/metabolismo , Utrofina/metabolismo
13.
BMC Vet Res ; 2: 12, 2006 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-16611357

RESUMEN

BACKGROUND: Degenerative suspensory ligament desmitis (DSLD) is a debilitating disorder thought to be limited to suspensory ligaments of Peruvian Pasos, Peruvian Paso crosses, Arabians, American Saddlebreds, American Quarter Horses, Thoroughbreds, and some European breeds. It frequently leads to persistent, incurable lameness and need to euthanize affected horses. The pathogenesis remains unclear, though the disease appears to run in families. Treatment and prevention are empirical and supportive, and not effective in halting the progression of the disease. Presently, the presumptive diagnosis of DSLD is obtained from patient signalment and history, clinical examination, and ultrasonographic examination of clinically affected horses, and is confirmed at post mortem examination. Presently, there are no reliable methods of diagnosing DSLD in asymptomatic horses. The goal of this study was to characterize and define the disorder in terms of tissue involvement at the macroscopic and microscopic levels. RESULTS: We examined tissues and organs from 28 affected horses (22 Peruvian Pasos, 6 horses of other breeds) and from 8 control horses. Histopathological examination revealed the presence of excessive amounts of proteoglycans in the following tissues removed from DSLD-affected horses: suspensory ligaments, superficial and deep digital flexor tendons, patellar and nuchal ligaments, cardiovascular system, and sclerae. Electron microscopy demonstrated changes in diameters of collagen fibrils in the tendon, and in smooth muscle cells of the media of the aorta compatible with increased cell permeability in DSLD-affected cells. Separation of tendon extracts by gel chromatography revealed the presence of additional proteoglycan(s) in extracts from affected, but not control extracts. CONCLUSION: This study demonstrates for the first time that DSLD, a disease process previously thought to be limited to the suspensory ligaments of the distal limbs of affected horses, is in fact a systemic disorder involving tissues and organs with significant connective tissue component. Abnormal accumulation of proteoglycans between collagen and elastic fibers rather than specific collagen fibril abnormalities is the most prominent histological feature of DSLD. Because of this observation and because of the involvement of many other tendons and ligaments beside the suspensory ligament, and of non-ligamentous tissue we, therefore, propose that equine systemic proteoglycan accumulation or ESPA rather than DSLD is a more appropriate name for this condition.

15.
Arch Toxicol ; 78(10): 599-608, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15148565

RESUMEN

Tendonitis and tendon rupture have been reported to occur during or following therapy with fluoroquinolone antibiotics. Though the pathogenesis is unknown, several studies suggest that fluoroquinolone antibiotics alter proteoglycan content in soft tissues, including tendons, and thereby alter collagen fibrillogenesis. To better understand the mechanism of action of fluoroquinolones, we studied the effects of enrofloxacin, a widely used fluoroquinolone in veterinary medicine, on avian tendon cell cultures established from gastrocnemius tendons from 18-day-old chicken embryos. We found that cell proliferation was progressively inhibited with increasing concentrations of enrofloxacin. This was accompanied by changes in morphology, extracellular matrix content and collagen fibril formation as detected by electron microscopy. We also observed a 35% decrease in the content of total monosaccharides in enrofloxacin-treated cells. The ratio of individual monosaccharides was also altered in enrofloxacin-treated cells. Enrofloxacin also induced the synthesis of small amounts of keratan sulfate in tendon cells. Moreover we observed enrofloxacin-induced changes in glycosylation of decorin, the most abundant tendon proteoglycan, resulting in the emergence of multiple lower molecular bands that were identifiable as decorin after chondroitinase ABC and N-glycanase treatment of extracts from enrofloxacin-treated cells. Medium conditioned by enrofloxacin-treated cells contained less decorin than did medium conditioned by control cells. We hypothesize that enrofloxacin induces either changes in the number of N-linked oligosaccharides attached to the core protein of decorin or changes in decorin degradation process. In conclusion, our data suggest that enrofloxacin affects cell proliferation and extracellular matrix through changes in glycosylation.


Asunto(s)
Antineoplásicos/toxicidad , Fluoroquinolonas/toxicidad , Glicosaminoglicanos/metabolismo , Proteoglicanos/metabolismo , Quinolonas/toxicidad , Tendones/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/metabolismo , Decorina , Relación Dosis-Respuesta a Droga , Enrofloxacina , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular , Cromatografía de Gases y Espectrometría de Masas , Sulfato de Queratano/biosíntesis , Músculo Esquelético , Proteoglicanos/análisis , Reticulina/efectos de los fármacos , Reticulina/ultraestructura , Tendones/metabolismo , Tendones/ultraestructura
16.
Anal Biochem ; 306(2): 298-304, 2002 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12123669

RESUMEN

The detection of microquantities of glycosaminoglycans (GAGs) in biological samples has been hampered by the lack of sensitive methods. In this paper we describe the modification and development of three sensitive assays capable of detecting nanogram quantities of GAGs in biological samples. The first assay detects total GAGs. It is a modified Alcian blue dye precipitation assay in which the dye binds to the negatively charged GAGs in CsCl-fractionated extracts from chicken tendons. This assay compares favorably with the widely used uronic acid assay in terms of its sensitivity and ability to detect all classes of GAGs, including keratan sulfate (KS). Two other assays, dot-blotting and immunoblotting, detect KS in complex mixtures and can be easily adapted for the detection of other GAGs. Both take advantage of binding of carboxyl and sulfate groups of GAGs to trivalent neodymium. In dot-blotting, samples were directly blotted onto nitrocellulose membrane soaked in Nd(2)(SO(4))(3) buffer, and KS was detected with the monoclonal anti-KS 5-D-4 antibody and an avidin-biotin complex detection system. In immunoblotting, the samples were first separated in 28% polyacrylamide gels, transferred onto a Nd(2)(SO(4))(3)-soaked nitrocellulose membrane using a phosphate buffer system, and stained and developed using the same protocol as in dot-blotting. Whereas dot-blotting allows the use of very low quantities of samples because of its high sensitivity (lower detection limit was 5 ng), immunoblotting provides more specificity.


Asunto(s)
Immunoblotting/métodos , Sulfato de Queratano/análisis , Azul Alcián/metabolismo , Animales , Pollos , Sulfato de Queratano/inmunología , Sulfato de Queratano/metabolismo , Ácidos Urónicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA