Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Angiogenesis ; 17(1): 77-91, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23959107

RESUMEN

Arterial and venous specification is critical for establishing and maintaining a functioning vascular system, and defects in key arteriovenous signaling pathways including VEGF (vascular endothelial growth factor) lead to congenital arteriopathies. The activities of VEGF, are in part controlled by heparan sulfate (HS) proteoglycans, significant components of the endothelial glycocalyx. The level of 6-O sulfation on HS polysaccharide chains, that mediate the interaction between HS and VEGFA, is edited at the cell surface by the enzyme SULF1. We investigated the role of sulf1 in vascular development. In zebrafish sulf1 is expressed in the head and tail vasculature, corresponding spatially and temporally with vascular development. Targeted knockdown of sulf1 by antisense morpholinos resulted in severe vascular patterning and maturation defects. 93 % of sulf1 morphants show dysmorphogenesis in arterial development leading to occlusion of the distal aorta and lack of axial and cranial circulation. Co-injection of vegfa165 mRNA rescued circulatory defects. While the genes affecting haematopoiesis are unchanged, expression of several arterial markers downstream of VegfA signalling such as notch and ephrinB2 are severely reduced in the dorsal aorta, with a concomitant increase in expression of the venous markers flt4 in the dorsal aorta of the morphants. Furthermore, in vitro, lack of SULF1 expression downregulates VEGFA-mediated arterial marker expression, confirming that Sulf1 mediates arterial specification by regulating VegfA165 activity. This study provides the first in vivo evidence for the integral role of the endothelial glycocalyx in specifying arterial-venous identity, vascular patterning and arterial integrity, and will help to better understand congenital arteriopathies.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Transducción de Señal/fisiología , Sulfatasas/biosíntesis , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Proteínas de Pez Cebra/biosíntesis , Pez Cebra/metabolismo , Animales , Arterias/embriología , Arterias/metabolismo , Efrina-B2/inmunología , Efrina-B2/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glicocálix/genética , Glicocálix/metabolismo , Morfolinos/farmacología , Oligonucleótidos Antisentido/farmacología , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Sulfatasas/antagonistas & inhibidores , Sulfatasas/genética , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Venas/embriología , Venas/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética
2.
Arterioscler Thromb Vasc Biol ; 31(3): 608-15, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21205989

RESUMEN

OBJECTIVE: Decorin and oxidized low-density lipoprotein (Ox-LDL) independently induce osteogenic differentiation of vascular smooth muscle cells (VSMCs). We aimed to determine whether decorin glycosaminoglycan (GAG) chain synthesis contributes to Ox-LDL-induced differentiation and calcification of human VSMCs in vitro. METHODS AND RESULTS: Human VSMCs treated with Ox-LDL to induce oxidative stress showed increased alkaline phosphatase (ALP) activity, accelerated mineralization, and a difference in both decorin GAG chain biosynthesis and CS/DS structure compared with untreated controls. Ox-LDL increased mRNA abundance of both xylosyltransferase (XT)-I, the key enzyme responsible for GAG chain biosynthesis and Msx2, a marker of osteogenic differentiation. Furthermore, downregulation of XT-I expression using small interfering RNA blocked Ox-LDL-induced VSMC mineralization. Adenoviral-mediated overexpression of decorin, but not a mutated unglycanated form, accelerated mineralization of VSMCs, suggesting GAG chain addition on decorin is crucial for the process of differentiation. The decorin-induced VSMC osteogenic differentiation involved activation of the transforming growth factor (TGF)-ß pathway, because it was attenuated by blocking of TGF-ß receptor signaling and because decorin overexpression potentiated phosphorylation of the downstream signaling molecule smad2. CONCLUSIONS: These studies provide direct evidence that oxidative stress-mediated decorin GAG chain synthesis triggers TGF-ß signaling and mineralization of VSMCs in vitro.


Asunto(s)
Calcinosis/metabolismo , Decorina/biosíntesis , Lipoproteínas LDL/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Osteogénesis , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Fosfatasa Alcalina/metabolismo , Células Cultivadas , Decorina/genética , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Estrés Oxidativo , Pentosiltransferasa/genética , Pentosiltransferasa/metabolismo , Fosforilación , Interferencia de ARN , Proteína Smad2/metabolismo , Factores de Tiempo , UDP Xilosa Proteína Xilosiltransferasa
3.
Am J Physiol Heart Circ Physiol ; 301(4): H1487-95, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21705677

RESUMEN

We investigated whether plasma long-chain sphingoid base (LCSB) concentrations are altered by transient cardiac ischemia during percutaneous coronary intervention (PCI) in humans and examined the signaling through the sphingosine-1-phosphate (S1P) cascade as a mechanism underlying the S1P cardioprotective effect in cardiac myocytes. Venous samples were collected from either the coronary sinus (n = 7) or femoral vein (n = 24) of 31 patients at 1 and 5 min and 12 h, following induction of transient myocardial ischemia during elective PCI. Coronary sinus levels of LCSB were increased by 1,072% at 1 min and 941% at 5 min (n = 7), while peripheral blood levels of LCSB were increased by 579% at 1 min, 617% at 5 min, and 436% at 12 h (n = 24). In cultured cardiac myocytes, S1P, sphingosine (SPH), and FTY720, a sphingolipid drug candidate, showed protective effects against CoCl induced hypoxia/ischemic cell injury by reducing lactate dehydrogenase activity. Twenty-five nanomolars of FTY720 significantly increased phospho-Pak1 and phospho-Akt levels by 56 and 65.6% in cells treated with this drug for 15 min. Further experiments demonstrated that FTY720 triggered nitric oxide release from cardiac myocytes is through pertussis toxin-sensitive phosphatidylinositol 3-kinase/Akt/endothelial nitric oxide synthase signaling. In ex vivo hearts, ischemic preconditioning was cardioprotective in wild-type control mice (Pak1(f/f)), but this protection appeared to be ineffective in cardiomyocyte-specific Pak1 knockout (Pak1(cko)) hearts. The present study provides the first direct evidence of the behavior of plasma sphingolipids following transient cardiac ischemia with dramatic and early increases in LCSB in humans. We also demonstrated that S1P, SPH, and FTY720 have protective effects against hypoxic/ischemic cell injury, likely a Pak1/Akt1 signaling cascade and nitric oxide release. Further study on a mouse model of cardiac specific deletion of Pak1 demonstrates a crucial role of Pak1 in cardiac protection against ischemia/reperfusion injury.


Asunto(s)
Cardiotónicos , Lisofosfolípidos/farmacología , Isquemia Miocárdica/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/fisiología , Proteína Oncogénica v-akt/fisiología , Esfingosina/análogos & derivados , Quinasas p21 Activadas/fisiología , Adulto , Anciano , Angioplastia Coronaria con Balón , Animales , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/fisiopatología , Biotransformación , Western Blotting , Hipoxia de la Célula/fisiología , Supervivencia Celular/fisiología , Oclusión Coronaria/metabolismo , Femenino , Clorhidrato de Fingolimod , Humanos , Técnicas In Vitro , L-Lactato Deshidrogenasa/metabolismo , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/fisiología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Proteína Oncogénica v-akt/genética , Toxina del Pertussis/farmacología , Glicoles de Propileno/farmacología , Ratas , Esfingolípidos/metabolismo , Esfingosina/farmacología , Quinasas p21 Activadas/genética
4.
Am J Pathol ; 177(4): 1946-57, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20813973

RESUMEN

Multiple osteochondromas (MO) is an autosomal dominant disorder caused by germline mutations in EXT1 and/or EXT2. In contrast, solitary osteochondroma (SO) is nonhereditary. Products of the EXT gene are involved in heparan sulfate (HS) biosynthesis. In this study, we investigated whether osteochondromas arise via either loss of heterozygosity (2 hits) or haploinsufficiency. An in vitro three-dimensional chondrogenic pellet model was used to compare heterozygous bone marrow-derived mesenchymal stem cells (MSCs EXT(wt/-)) of MO patients with normal MSCs and the corresponding tumor specimens (presumed EXT(-/-)). We demonstrated a second hit in EXT in five of eight osteochondromas. HS chain length and structure, in vitro chondrogenesis, and EXT expression levels were identical in both EXT(wt/-) and normal MSCs. Immunohistochemistry for HS, HS proteoglycans, and HS-dependent signaling pathways (eg, TGF-ß/BMP, Wnt, and PTHLH) also showed no differences. The cartilaginous cap of osteochondroma contained a mixture of HS-positive and HS-negative cells. Because a heterozygous EXT mutation does not affect chondrogenesis, EXT, HS, or downstream signaling pathways in MSCs, our results refute the haploinsufficiency theory. We found a second hit in 63% of analyzed osteochondromas, supporting the hypothesis that osteochondromas arise via loss of heterozygosity. The detection of the second hit may depend on the ratio of HS-positive (normal) versus HS-negative (mutated) cells in the cartilaginous cap of the osteochondroma.


Asunto(s)
Exostosis Múltiple Hereditaria/genética , Haploinsuficiencia/genética , Pérdida de Heterocigocidad/genética , N-Acetilglucosaminiltransferasas/genética , Adolescente , Adulto , Western Blotting , Médula Ósea/metabolismo , Estudios de Casos y Controles , Diferenciación Celular , Células Cultivadas , Niño , Femenino , Citometría de Flujo , Mutación de Línea Germinal/genética , Proteoglicanos de Heparán Sulfato/metabolismo , Heparitina Sulfato/metabolismo , Heterocigoto , Humanos , Técnicas para Inmunoenzimas , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Persona de Mediana Edad , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factor de Crecimiento Transformador beta
5.
Dev Dyn ; 239(12): 3312-23, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20981828

RESUMEN

The 6-O-endosulfatase enzymes (Sulfs) edit the final sulfation pattern and function of heparan sulfate (HS) by removal of 6-O-sulfate groups from the chain. To date, two mammalian sulf genes have been identified that regulate many signalling pathways during embryonic development. In zebrafish a sulf1 ortholog and duplicate copies of the mammalian sulf2 gene, sulf2a and sulf2, have been identified, which contain conserved motifs characteristic of vertebrate sulf genes. Zebrafish sulf1 and sulf2a are broadly expressed in the central nervous system (CNS) and non-neuronal tissue including heart, somite boundaries, olfactory system, and otic vesicle, whereas sulf2 expression is almost entirely restricted to the CNS. The duplicate copies of sulf2 have distinct expression patterns, which together mirror that of mouse sulf2, suggesting duplication in the teleost lineage has been followed by subfunctionalisation, whereby both genes need to be preserved by selection to ensure the ancestral gene's expression profile and function is maintained.


Asunto(s)
Proteínas de Pez Cebra/metabolismo , Animales , Sistema Nervioso Central/embriología , Biología Computacional , Embrión no Mamífero/metabolismo , Corazón/embriología , Heparitina Sulfato/metabolismo , Hibridación in Situ , Vías Olfatorias/embriología , Proteoglicanos/metabolismo , Proteínas de Pez Cebra/genética
6.
Neuron ; 44(6): 947-60, 2004 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-15603738

RESUMEN

Retinal ganglion cell (RGC) axons are topographically ordered in the optic tract according to their retinal origin. In zebrafish dackel (dak) and boxer (box) mutants, some dorsal RGC axons missort in the optic tract but innervate the tectum topographically. Molecular cloning reveals that dak and box encode ext2 and extl3, glycosyltransferases implicated in heparan sulfate (HS) biosynthesis. Both genes are required for HS synthesis, as shown by biochemical and immunohistochemical analysis, and are expressed maternally and then ubiquitously, likely playing permissive roles. Missorting in box can be rescued by overexpression of extl3. dak;box double mutants show synthetic pathfinding phenotypes that phenocopy robo2 mutants, suggesting that Robo2 function requires HS in vivo; however, tract sorting does not require Robo function, since it is normal in robo2 null mutants. This genetic evidence that heparan sulfate proteoglycan function is required for optic tract sorting provides clues to begin understanding the underlying molecular mechanisms.


Asunto(s)
Axones/metabolismo , Proteoglicanos de Heparán Sulfato/biosíntesis , N-Acetilglucosaminiltransferasas/fisiología , Vías Visuales/metabolismo , Proteínas de Pez Cebra/fisiología , Animales , Proteoglicanos de Heparán Sulfato/genética , Datos de Secuencia Molecular , N-Acetilglucosaminiltransferasas/biosíntesis , N-Acetilglucosaminiltransferasas/genética , Células Ganglionares de la Retina/metabolismo , Vías Visuales/embriología , Pez Cebra
7.
Int J Biochem Cell Biol ; 35(7): 1052-5, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12672474

RESUMEN

The CXC chemokine platelet factor 4 (PF4) appears to inhibit tumour growth through its modulation of the activity of angiogenic growth factors. We investigated the heparan sulphate-dependent mechanism of PF4 inhibition of fibroblast growth factor 2 (FGF-2). The ability of PF4 to bind simultaneously to both FGF-2 and HS was assessed using affinity gel chromatography. Thirty-three to forty-two percent more HS bound to the FGF-2 affinity gel in the presence of PF4 than with HS alone. Protection assays showed that PF4 and FGF-2 bound to adjacent or overlapping sites together covering a 12 kDa stretch of HS. This study suggests that the three components may form a ternary complex. PF4 released at sites of angiogenesis may bind to angiogenic growth factors attached to endothelial cell surface HS to disrupt or prevent them from interacting with their signalling receptors. Manipulation of this mechanism may prove useful for clinical intervention of angiogenesis.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Heparitina Sulfato/metabolismo , Factor Plaquetario 4/metabolismo , Células 3T3 , Animales , Sitios de Unión , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Técnicas In Vitro , Ratones
8.
Aging Cell ; 12(1): 139-47, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23190312

RESUMEN

Aging poses one of the largest risk factors for the development of cardiovascular disease. The increased propensity toward vascular pathology with advancing age maybe explained, in part, by a reduction in the ability of circulating endothelial progenitor cells to contribute to vascular repair and regeneration. Although there is evidence to suggest that colony forming unit-Hill cells and circulating angiogenic cells are subject to age-associated changes that impair their function, the impact of aging on human outgrowth endothelial cell (OEC) function has been less studied. We demonstrate that OECs isolated from cord blood or peripheral blood samples from young and old individuals exhibit different characteristics in terms of their migratory capacity. In addition, age-related structural changes were discovered in OEC heparan sulfate (HS), a glycocalyx component that is essential in many signalling pathways. An age-associated decline in the migratory response of OECs toward a gradient of VEGF significantly correlated with a reduction in the relative percentage of the trisulfated disaccharide, 2-O-sulfated-uronic acid, N, 6-O-sulfated-glucosamine (UA[2S]-GlcNS[6S]), within OEC cell surface HS polysaccharide chains. Furthermore, disruption of cell surface HS reduced the migratory response of peripheral blood-derived OECs isolated from young subjects to levels similar to that observed for OECs from older individuals. Together these findings suggest that aging is associated with alterations in the fine structure of HS on the cell surface of OECs. Such changes may modulate the migration, homing, and engraftment capacity of these repair cells, thereby contributing to the progression of endothelial dysfunction and age-related vascular pathologies.


Asunto(s)
Movimiento Celular/fisiología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Células Madre/citología , Células Madre/metabolismo , Factores de Edad , Apoptosis/fisiología , Células Sanguíneas/citología , Procesos de Crecimiento Celular/fisiología , Supervivencia Celular/fisiología , Sangre Fetal/citología , Humanos , Longevidad
9.
Front Physiol ; 4: 130, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23781203

RESUMEN

BACKGROUND: Ventricular tachyarrhythmias are the most common and often the first manifestation of coronary heart disease and lead to sudden cardiac death (SCD). Early detection/identification of acute myocardial ischaemic injury at risk for malignant ventricular arrhythmias in patients remains an unmet medical need. In the present study, we examined the sphingolipids level after transient cardiac ischaemia following temporary coronary artery occlusion during percutaneous coronary intervention (PCI) in patients and determined the role of sphingolipids level as a novel marker for early detection of human myocardial ischaemic injury. METHODS AND RESULTS: Venous samples were collected from either the coronary sinus (n = 7) or femoral vein (n = 24) from 31 patients aged 40-73 years-old at 1, 5 min, and 12 h, following elective PCI. Plasma sphingolipids levels were assessed by HPLC. At 1 min coronary sinus levels of sphingosine 1-phosphate (S1P), sphingosine (SPH), and sphinganine (SA) were increased by 314, 115, and 614%, respectively (n = 7), while peripheral blood levels increased by 79, 68, and 272% (n = 24). By 5 min, coronary sinus S1P and SPH levels increased further (720%, 117%), as did peripheral levels of S1P alone (792%). Where troponin T was detectable at 12 h (10 of 31), a strong correlation was found with peak S1P (R (2) = 0.818; P < 0.0001). CONCLUSION: For the first time, we demonstrate the behavior of plasma sphingolipids following transient cardiac ischaemia in humans. The observation supports the important role of sphingolipids level as a potential novel marker of transient or prolonged myocardial ischaemia.

10.
ACS Chem Biol ; 8(10): 2331-8, 2013 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-23972127

RESUMEN

Heparan sulfate (HS), a long linear polysaccharide, is implicated in various steps of tumorigenesis, including angiogenesis. We successfully interfered with HS biosynthesis using a peracetylated 4-deoxy analogue of the HS constituent GlcNAc and studied the compound's metabolic fate and its effect on angiogenesis. The 4-deoxy analogue was activated intracellularly into UDP-4-deoxy-GlcNAc, and HS expression was inhibited up to ∼96% (IC50 = 16 µM). HS chain size was reduced, without detectable incorporation of the 4-deoxy analogue, likely due to reduced levels of UDP-GlcNAc and/or inhibition of glycosyltransferase activity. Comprehensive gene expression analysis revealed reduced expression of genes regulated by HS binding growth factors such as FGF-2 and VEGF. Cellular binding and signaling of these angiogenic factors was inhibited. Microinjection in zebrafish embryos strongly reduced HS biosynthesis, and angiogenesis was inhibited in both zebrafish and chicken model systems. All of these data identify 4-deoxy-GlcNAc as a potent inhibitor of HS synthesis, which hampers pro-angiogenic signaling and neo-vessel formation.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Heparitina Sulfato/genética , Neovascularización Patológica/fisiopatología , Uridina Difosfato N-Acetilglucosamina/análogos & derivados , Uridina Difosfato N-Acetilglucosamina/farmacología , Animales , Pollos , Regulación hacia Abajo/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/genética , Heparitina Sulfato/biosíntesis , Heparitina Sulfato/metabolismo , Ácido Idurónico/química , Transducción de Señal/efectos de los fármacos , Uridina Difosfato N-Acetilglucosamina/química , Uridina Difosfato N-Acetilglucosamina/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Pez Cebra
11.
Virchows Arch ; 461(4): 475-81, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22903264

RESUMEN

Chondrosarcomas are malignant cartilage-forming tumours that can arise centrally (in the medulla) or peripherally (at the surface) of the bone. They are classified into three histological grades which correspond to the clinical severity. Previous studies by our group have shown altered signal transduction of the fibroblast growth factor and Wnt signalling pathways during peripheral chondrosarcoma progression. Heparan sulphate (HS) is a glycosaminoglycan that facilitates receptor binding of multiple growth factors, in which the sulphation of 6O position plays a pivotal role. 6O-Sulphation occurs through three HS 6O-sulphotransferases (HS6ST1-3) and is fine-tuned by two endosulphatases (SULF1-2) that remove 6O-sulphate groups. We have investigated whether the expression of HS6STs and SULFs changes during chondrosarcoma progression and have determined 6O-sulphation levels in two chondrosarcoma cell lines. Immunohistochemistry on tissue microarrays of chondrosarcomas showed that HS6ST3 and SULF1 were highly expressed in most chondrosarcomas, whereas SULF2 expression was absent in most cases. HS6ST1 and HS6ST2 expression are significantly increased during chondrosarcoma progression, which suggest that 6O-sulphation is increased during progression. This was confirmed in one grade III chondrosarcoma cell line, which showed a dramatically increased 6O-sulphation compared to an articular chondrocyte cell line by HPLC; another cell line showed an increased expression of one 6O-sulphated HS disaccharide. In conclusion, our results show increased HS6ST1 and HS6ST2 expression during chondrosarcoma progression and increased HS 6O-sulphation in vitro. As 6O-sulphation plays an important role in signal transduction, altered HS6ST expression might be associated with changes in signal transduction pathways in chondrosarcoma progression.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/metabolismo , Condrosarcoma/metabolismo , Progresión de la Enfermedad , Sulfotransferasas/metabolismo , Biopsia , Neoplasias Óseas/patología , Línea Celular Tumoral , Células Cultivadas , Condrocitos/metabolismo , Condrocitos/patología , Condrosarcoma/patología , Factores de Crecimiento de Fibroblastos/metabolismo , Estudios de Seguimiento , Humanos , Técnicas In Vitro , Análisis por Micromatrices , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo
12.
Trends Cell Biol ; 17(4): 173-7, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17320398

RESUMEN

Heparan sulfate (HS) proteoglycans, at the cell surface and extracellular matrix, facilitate ligand-receptor interactions crucial to many physiological processes. The distinct sulfation patterns of HS sugar chains presented by their protein core are key to HS proteoglycan activity. Tight regulation of several Golgi complex enzyme families is crucial to produce complex tissue-specific HS sequences. Several in vivo models deficient in HS biosynthesis enzymes demonstrate that developmental abnormalities result from modified HS structure. This review will discuss the plasticity of sulfation requirements on HS for activating protein ligands, which might reflect a flexible HS biosynthetic mechanism. In addition, the latest discovery of HS acting enzymes, the Sulfs, responsible for extracellular tweaking of HS sulfation levels subsequent to biosynthesis will be considered.


Asunto(s)
Heparitina Sulfato/metabolismo , Morfogénesis/fisiología , Animales , Carbohidrato Epimerasas/metabolismo , Heparitina Sulfato/biosíntesis , Humanos , N-Acetilglucosaminiltransferasas/genética , Sulfatasas/metabolismo , Sulfotransferasas/genética
13.
J Biol Chem ; 281(3): 1731-40, 2006 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-16258170

RESUMEN

The vascular endothelial growth factor (VEGF) family of proteins controls the formation and growth of blood vessels. The most potent and widely expressed isoform, VEGF165, is secreted as a disulfide-linked homodimer with two identical heparin-binding sites. Interactions with heparan sulfate (HS) regulate the diffusion, half-life, and affinity of VEGF165 for its signaling receptors. We have determined a number of key HS structural features that mediate the specific binding of the VEGF165 dimer. Carboxylate groups and 2-O-, 6-O-, and N-sulfation of HS contributed to the strength of the VEGF165 interaction; however, 6-O-sulfates appeared to be particularly important. Cleavage of HS by heparinase, heparitinase, or heparanase severely reduced VEGF165 binding. In contrast, K5 lyase-cleaved HS retained significant VEGF165 affinity, suggesting that binding sites for the growth factor are present within extended stretches of sulfation. Binding studies and molecular modeling demonstrated that an oligosaccharide 6 or 7 residues long was sufficient to fully occupy the heparin-binding site of a VEGF165 monomer. The data presented are consistent with a model whereby the two heparin-binding sites of the VEGF165 dimer interact simultaneously with highly sulfated S-domain regions of the HS chain that can be linked through a stretch of transition sequence.


Asunto(s)
Heparitina Sulfato/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células 3T3 , Animales , Conformación de Carbohidratos , Bovinos , Cromatografía Líquida de Alta Presión , Dimerización , Disacáridos/química , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/química , Heparitina Sulfato/aislamiento & purificación , Ratones , Modelos Moleculares , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Tiburones , Factor A de Crecimiento Endotelial Vascular/química
14.
Dev Biol ; 284(2): 364-76, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16009360

RESUMEN

Heparan sulfate proteoglycans are important modulators of growth factor signaling in a variety of patterning processes. Secreted growth factors that play critical roles in angiogenesis bind to heparan sulfate, and this association is affected by 6-O-sulfation of the heparan sulfate chains. Addition of 6-O-sulfate is catalyzed by a family of sulfotransferases (HS6STs), and genetic manipulation of their function permits an assessment of their contribution to vascular assembly. We report on the biochemical activity and expression patterns of two zebrafish HS6ST genes. In situ hybridization reveals dynamic and distinct expression patterns of these two genes during development. Structural analysis of heparan sulfate from wild-type and morpholino antisense 'knockdown' embryos suggests that HS6ST-1 and HS6ST-2 have similar biochemical activity. HS6ST-2, but not HS6ST-1, morphants exhibit abnormalities in the branching morphogenesis of the caudal vein during embryonic development of the zebrafish. Our finding that HS6ST-2 is required for the branching morphogenesis of the caudal vein is the first in vivo evidence for an essential role of a gene encoding a heparan sulfate modifying enzyme in vertebrate angiogenesis. Our analysis of two zebrafish HS6ST genes suggests that a wide range of biological processes may be regulated by an array of sulfation-modifying enzymes in the vertebrate genome.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Sulfotransferasas/metabolismo , Pez Cebra/embriología , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Vasos Sanguíneos/embriología , Clonación Molecular , Embrión no Mamífero , Desarrollo Embrionario , Etiquetas de Secuencia Expresada , Proteínas Fluorescentes Verdes/metabolismo , Heparitina Sulfato/metabolismo , Hibridación in Situ , Microinyecciones , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/farmacología , Filogenia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Somitos/metabolismo , Sulfotransferasas/antagonistas & inhibidores , Sulfotransferasas/química , Sulfotransferasas/genética , Sulfotransferasas/aislamiento & purificación
15.
Blood ; 106(6): 1956-64, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15947088

RESUMEN

In mucopolysaccharidosis-I (MPS-I), alpha-L-iduronidase deficiency leads to progressive heparan sulfate (HS) and dermatan sulfate (DS) glycosaminoglycan (GAG) accumulation. The functional consequences of these accumulated molecules are unknown. HS critically influences tissue morphogenesis by binding to and modulating the activity of several cytokines (eg, fibroblast growth factors [FGFs]) involved in developmental patterning. We recently isolated a multipotent progenitor cell from postnatal human bone marrow, which differentiates into cells of all 3 embryonic lineages. The availability of multipotent progenitor cells from healthy volunteers and patients with MPS-I (Hurler syndrome) provides a unique opportunity to directly examine the functional effects of abnormal HS on cytokine-mediated stem-cell proliferation and survival. We demonstrate here that abnormally sulfated HS in Hurler multipotent progenitor cells perturb critical FGF-2-FGFR1-HS interactions, resulting in defective FGF-2-induced proliferation and survival of Hurler multipotent progenitor cells. Both the mitogenic and survival-promoting activities of FGF-2 were restored by substitution of Hurler HS by normal HS. This perturbation of critical HS-cytokine receptor interactions may represent a mechanism by which accumulated HS contributes to the developmental pathophysiology of Hurler syndrome. Similar mechanisms may operate in the pathogenesis of other diseases where structurally abnormal GAGs accumulate.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Heparitina Sulfato/química , Heparitina Sulfato/fisiología , Mucopolisacaridosis I/metabolismo , Células Madre Multipotentes/patología , Estudios de Casos y Controles , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Cromatografía Líquida de Alta Presión , Heparitina Sulfato/aislamiento & purificación , Humanos , Mucopolisacaridosis I/etiología , Células Madre Multipotentes/citología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo
16.
Blood ; 101(6): 2243-5, 2003 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-12406885

RESUMEN

We previously showed that heparan sulfate (HS) is required for in vitro cytokine + chemokine-mediated maintenance of primitive human hematopoietic progenitors. However, HS preparations are mixtures of polysaccharide chains of varying size, structure, and protein-binding abilities. Therefore, we examined whether the long-term culture-initiating cells (LTC-IC) supportive capability of HS is attributable to an oligosaccharide of defined length and protein-binding ability. Oligosaccharides of a wide range of sizes were prepared, and their capability to support human marrow LTC-IC maintenance in the presence of low-dose cytokines and a single chemokine, macrophage inflammatory protein-1alpha (MIP-1alpha), was examined. LTC-IC supportive capability of HS oligosaccharides correlated directly with size and MIP-1alpha binding ability. A specific MIP-1alpha-binding HS oligosaccharide preparation of M(r) 10 kDa that optimally supported LTC-IC maintenance was identified. This oligosaccharide had the structure required for MIP-1alpha binding, which we have recently described. The present study defines the minimum size and structural features of LTC-IC supportive HS.


Asunto(s)
Células Madre Hematopoyéticas/citología , Heparitina Sulfato/química , Proteínas Inflamatorias de Macrófagos/metabolismo , Oligosacáridos/metabolismo , Oligosacáridos/farmacología , Antígenos CD34/análisis , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células Cultivadas , Quimiocina CCL3 , Quimiocina CCL4 , Antígenos HLA-DR/análisis , Células Madre Hematopoyéticas/inmunología , Heparitina Sulfato/metabolismo , Humanos , Oligosacáridos/química , Unión Proteica
17.
Glycobiology ; 13(2): 97-107, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12626406

RESUMEN

Heparin is a highly sulfated glycosaminoglycan widely used as an anticoagulant. Modifications in its relatively uniform structure appear to be key to its recognition and modulation of serine proteases, growth factors, chemokines, and extracellular proteins, as has been most clearly demonstrated in the antithrombin binding site. We sequenced the major oligosaccharides released from mastocytoma heparin by partial nitrous acid using a highly sensitive technique tailored for sequencing of metabolically radiolabeled heparin. It utilizes partial nitrous acid cleavage to allow simultaneous sequencing of the internal components of the oligosaccharide under investigation by specific lysosomal exoenzymes. Sequencing revealed that although the majority of the heparin disaccharides are N-, 2-O-, and 6-O-sulfated, the less sulfated disaccharides (lacking 2-O- or 6-O-sulfates) seem to be spaced out along the chain. The technique may be particularly useful for characterizing heparin from novel sources, such as the glial progenitor cells and Ascidia, as well as for sequencing protein binding sites.


Asunto(s)
Heparina/análisis , Análisis de Secuencia/métodos , Animales , Secuencia de Carbohidratos , Cromatografía Líquida de Alta Presión/métodos , Electroforesis en Gel de Poliacrilamida/métodos , Iduronidasa/metabolismo , Datos de Secuencia Molecular , Ácido Nitroso/metabolismo , Oligosacáridos/aislamiento & purificación , Oligosacáridos/metabolismo , Tamaño de la Partícula , Sulfatasas/metabolismo , Células Tumorales Cultivadas
18.
Blood ; 100(5): 1543-50, 2002 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12176868

RESUMEN

The CC chemokine macrophage inflammatory protein 1alpha (MIP1alpha) is a key regulator of the proliferation and differentiation of hematopoietic progenitor cells. The activity of MIP1alpha appears to be modulated by its binding to heparan sulfate (HS) proteoglycans, ubiquitous components of the mammalian cell surface and extracellular matrix. In this study we show that HS has highest affinity for the dimeric form of MIP1alpha. The predominantly dimeric BB10010 MIP1alpha interacts with an 8.3-kDa sequence in the HS polysaccharide chain, which it protects from degradation by heparinase enzymes. The major structural motif of this HS fragment appears to consist of 2 sulfate-rich S-domains separated by a short central N-acetylated region. The optimum lengths of these S-domains seem to be 12 to 14 saccharides. We propose that this binding fragment may wrap around the MIP1alpha dimer in a horseshoe shape, facilitating the interaction of the S-domains with the heparin-binding domains on each monomer. Molecular modeling suggests that these S-domains are likely to interact with basic residues Arg 17, Arg 45, and Arg 47 and possibly with Lys 44 on MIP1alpha and that the interconnecting N-acetylated region is of sufficient length to allow the 2 S-domains to bind to these sites on opposite faces of the dimer. Elucidation of the structure of the HS-binding site for MIP1alpha may enable us to devise ways of enhancing its myeloprotective or peripheral blood stem cell mobilization properties, which can be used to improve cancer chemotherapy treatments.


Asunto(s)
Heparitina Sulfato/metabolismo , Proteínas Inflamatorias de Macrófagos/metabolismo , Células del Estroma/metabolismo , Sitios de Unión , Quimiocina CCL4 , Dimerización , Heparitina Sulfato/química , Humanos , Técnicas In Vitro , Proteínas Inflamatorias de Macrófagos/química , Unión Proteica , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA