Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Glycobiology ; 31(11): 1531-1542, 2021 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-34324645

RESUMEN

Fractones, specialized extracellular matrix structures found in the subventricular zone (SVZ) neurogenic niche, can capture growth factors, such as basic fibroblast growth factor, from the extracellular milieu through a heparin-binding mechanism for neural stem cell (NSC) presentation, which promotes neurogenesis. During aging, a decline in neurogenesis correlates with a change in the composition of heparan sulfate (HS) within fractones. In this study, we used antibodies that recognize specific short oligosaccharides with varying sulfation to evaluate the HS composition in fractones in young and aged brains. To further understand the conditions that regulate 6-O sulfation levels and its impact on neurogenesis, we used endosulfatase Sulf1 and Sulf2 double knockout (DKO) mice. Fractones in the SVZ of Sulf1/2 DKO mice showed immunoreactivity for the HS epitope, suggesting higher 6-O sulfation. While neurogenesis declined in the aged SVZ of both wild-type and Sulf1/2 DKO mice, we observed a larger number of neuroblasts in the young and aged SVZ of Sulf1/2 DKO mice. Together, these results show that the removal of 6-O-sulfation in fractones HS by endosulfatases inhibits neurogenesis in the SVZ. Our findings advance the current understanding regarding the extracellular environment that is best suited for NSCs to thrive, which is critical for the design of future stem cell therapies.


Asunto(s)
Heparitina Sulfato/metabolismo , Ventrículos Laterales/metabolismo , Sulfatasas/metabolismo , Sulfotransferasas/metabolismo , Animales , Matriz Extracelular , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Neurogénesis , Nicho de Células Madre , Sulfatasas/deficiencia , Sulfotransferasas/deficiencia
2.
Dev Neurosci ; 39(5): 361-374, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28490013

RESUMEN

Sulfatases (Sulfs) are a group of endosulfatases consisting of Sulf1 and Sulf2, which specifically remove sulfate from heparan sulfate proteoglycans. Although several studies have shown that Sulf1 acts as a regulator of sonic hedgehog (Shh) signaling during embryonic ventral spinal cord development, the detailed expression pattern and function of Sulf2 in the spinal cord remains to be determined. In this study, we found that Sulf2 also modulates the cell fate change from motor neurons (MNs) to oligodendrocyte precursor cells (OPCs) by regulating Shh signaling in the mouse ventral spinal cord in coordination with Sulf1. In the mouse, Sulf mRNAs colocalize with Shh mRNA and gradually expand dorsally from embryonic day (E) 10.5 to E12.5, following strong Patched1 signals (a target gene of Shh signaling). This coordinated expression pattern led us to hypothesize that in the mouse, strong Shh signaling is induced when Shh is released by Sulf1/2, and this strong Shh signaling subsequently induces the dorsal expansion of Shh and Sulf1/2 expression. Consistent with this hypothesis, in the ventral spinal cord of Sulf1 knockout (KO) or Sulf2 KO mice, the expression patterns of Shh and Patched1 differed from that in wild-type mice. Moreover, the position of the pMN and p3 domains were shifted ventrally, MN generation was prolonged, and OPC generation was delayed at E12.5 in both Sulf1 KO and Sulf2 KO mice. These results demonstrated that in addition to Sulf1, Sulf2 also plays an important and overlapping role in the MN-to-OPC fate change by regulating Shh signaling in the ventral spinal cord. However, neither Sulf1 nor Sulf2 could compensate for the loss of the other in the developing mouse spinal cord. In vitro studies showed no evidence of an interaction between Sulf1 and Sulf2 that could increase sulfatase activity. Furthermore, Sulf1/2 double heterozygote and Sulf1/2 double KO mice exhibited phenotypes similar to the Sulf1 KO and Sulf2 KO mice. These results indicate that there is a threshold for sulfatase activity (which is likely reflected in the dose of Shh) required to induce the MN-to-OPC fate change, and Shh signaling requires the coordinated activity of Sulf1 and Sulf2 in order to reach that threshold in the mouse ventral spinal cord.


Asunto(s)
Proteínas Hedgehog/metabolismo , Neuronas Motoras/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Transducción de Señal , Sulfatasas/metabolismo , Sulfotransferasas/metabolismo , Animales , Diferenciación Celular/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/fisiología , Médula Espinal/metabolismo , Sulfatasas/genética , Sulfotransferasas/genética
3.
Am J Physiol Renal Physiol ; 310(5): F395-408, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26764203

RESUMEN

Glomerular integrity and functions are maintained by growth factor signaling. Heparan sulfate, the major component of glomerular extracellular matrixes, modulates growth factor signaling, but its roles in glomerular homeostasis are unknown. We investigated the roles of heparan sulfate 6-O-endosulfatases, sulfatase (Sulf)1 and Sulf2, in glomerular homeostasis. Both Sulf1 and Sulf2 were expressed in the glomeruli of wild-type (WT) mice. Sulf1 and Sulf2 double-knockout (DKO) mice showed glomerular hypercellularity, matrix accumulation, mesangiolysis, and glomerular basement membrane irregularity. Platelet-derived growth factor (PDGF)-B and PDGF receptor-ß were upregulated in Sulf1 and Sulf2 DKO mice compared with WT mice. Glomeruli from Sulf1 and Sulf2 DKO mice in vitro stimulated by either PDGF-B, VEGF, or transforming growth factor-ß similarly showed reduction of phospho-Akt, phospho-Erk1/2, and phospho-Smad2/3, respectively. Since glomerular lesions in Sulf1 and Sulf2 DKO mice were reminiscent of diabetic nephropathy, we examined the effects of Sulf1 and Sulf2 gene disruption in streptozotocin-induced diabetes. Diabetic WT mice showed an upregulation of glomerular Sulf1 and Sulf2 mRNA by in situ hybridization. Diabetic DKO mice showed significant increases in albuminuria and serum creatinine and an acceleration of glomerular pathology without glomerular hypertrophy; those were associated with a reduction of glomerular phospho-Akt. In conclusion, Sulf1 and Sulf2 play indispensable roles to maintain glomerular integrity and protective roles in diabetic nephropathy, probably by growth factor modulation.


Asunto(s)
Nefropatías Diabéticas/enzimología , Heparitina Sulfato/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Glomérulos Renales/efectos de los fármacos , Receptores de Factores de Crecimiento/agonistas , Transducción de Señal/efectos de los fármacos , Sulfatasas/metabolismo , Sulfotransferasas/metabolismo , Animales , Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Predisposición Genética a la Enfermedad , Glomérulos Renales/enzimología , Glomérulos Renales/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , Receptores de Factores de Crecimiento/metabolismo , Proteínas Smad Reguladas por Receptores/metabolismo , Sulfatasas/deficiencia , Sulfatasas/genética , Sulfotransferasas/deficiencia , Sulfotransferasas/genética , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Factor de Crecimiento Transformador beta/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología
4.
J Neurochem ; 139 Suppl 2: 58-75, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26709493

RESUMEN

Neural circuits are formed with great precision during development. Accumulated evidence over the past three decades has demonstrated that growing axons are navigated toward their targets by the combined actions of attractants and repellents together with their receptors. It has long been known that proteoglycans, glycosylated proteins possessing covalently attached glycosaminoglycans, play a critical role in axon guidance; however, the molecular mechanisms by which proteoglycans regulate axon behaviors remain largely unknown. Glycosaminoglycans such as heparan sulfate and chondroitin sulfate are large linear polysaccharides composed of repeating disaccharide units that are highly modified by specific sulfation and epimerization. Recent biochemical and molecular biological studies have identified the enzymes that are involved in the biosynthesis of glycosaminoglycans. Interestingly, many mutants lacking glycosaminoglycan-synthesizing enzymes or proteoglycans in several model organisms show defects in specific nerve tract formation. In parallel, detailed biochemical studies have identified the molecular interactions between axon guidance molecules and glycosaminoglycans that have specific modification in their sugar chains. This review summarizes the structure and function of axon guidance molecules and glycosaminoglycans, and then tries to combine the knowledge from these studies to understand the role of proteoglycans from a new vantage point. Deciphering the sugar code is important for understanding the complicated nature of proteoglycans in axon guidance. Neural circuits are formed by the combined actions of axon guidance molecules. Proteoglycans play critical roles in regulating axon guidance through the interaction between signaling molecules and glycosaminoglycan chains attached to the core protein. This paper summarizes the structure and functions of axon guidance molecules and glycosaminoglycans and reviews the molecular mechanisms by which proteoglycans regulate axon guidance from a new vantage point. This article is part of the 60th Anniversary special issue.


Asunto(s)
Orientación del Axón/fisiología , Axones/metabolismo , Proteoglicanos/metabolismo , Animales , Glicosaminoglicanos/metabolismo , Humanos , Regeneración Nerviosa/fisiología
5.
Dev Dyn ; 244(2): 168-80, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25370455

RESUMEN

BACKGROUND: Inner ear morphogenesis is tightly regulated by the temporally and spatially coordinated action of signaling ligands and their receptors. Ligand-receptor interactions are influenced by heparan sulfate proteoglycans (HSPGs), cell surface molecules that consist of glycosaminoglycan chains bound to a protein core. Diversity in the sulfation pattern within glycosaminoglycan chains creates binding sites for numerous cell signaling factors, whose activities and distribution are modified by their association with HSPGs. RESULTS: Here we describe the expression patterns of two extracellular 6-O-endosulfatases, Sulf1 and Sulf2, whose activity modifies the 6-O-sulfation pattern of HSPGs. We use in situ hybridization to determine the temporal and spatial distribution of transcripts during the development of the chick and mouse inner ear. We also use immunocytochemistry to determine the cellular localization of Sulf1 and Sulf2 within the sensory epithelia. Furthermore, we analyze the organ of Corti in Sulf1/Sulf2 double knockout mice and describe an increase in the number of mechanosensory hair cells. CONCLUSIONS: Our results suggest that the tuning of intracellular signaling, mediated by Sulf activity, plays an important role in the development of the inner ear.


Asunto(s)
Proteínas Aviares/biosíntesis , Regulación del Desarrollo de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Órgano Espiral/embriología , Sulfatasas/biosíntesis , Sulfotransferasas/biosíntesis , Animales , Embrión de Pollo , Ratones , Órgano Espiral/citología , Transducción de Señal/fisiología
6.
MicroPubl Biol ; 20242024.
Artículo en Inglés | MEDLINE | ID: mdl-38725939

RESUMEN

Autotaxin, encoded by the Enpp2 gene, produces lysophosphatidic acid (LPA), which exerts numerous biological functions via its cognate receptors. Enpp2 null mutant mice die by embryonic day 9.5 owing to aberrant vascular development in the yolk sac, preventing analysis after that period. In this study, we found that Enpp2 heterozygous mice in the DBA/2 genetic background showed an eye-open-at-birth phenotype at high frequency, caused by failure of eyelid closure during the embryonic stage. Notably, wildtype pups from the Enpp2 heterozygous dam showed the phenotype, although at lower frequency, suggesting that maternal LPA affects the embryonic development.

7.
J Biol Chem ; 287(12): 9579-90, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22298771

RESUMEN

Heparan sulfate endosulfatases Sulf1 and Sulf2 hydrolyze 6-O-sulfate in heparan sulfate, thereby regulating cellular signaling. Previous studies have revealed that Sulfs act predominantly on UA2S-GlcNS6S disaccharides and weakly on UA-GlcNS6S disaccharides. However, the specificity of Sulfs and their role in sulfation patterning of heparan sulfate in vivo remained unknown. Here, we performed disaccharide analysis of heparan sulfate in Sulf1 and Sulf2 knock-out mice. Significant increases in ΔUA2S-GlcNS6S were observed in the brain, small intestine, lung, spleen, testis, and skeletal muscle of adult Sulf1(-/-) mice and in the brain, liver, kidney, spleen, and testis of adult Sulf2(-/-) mice. In addition, increases in ΔUA-GlcNS6S were seen in the Sulf1(-/-) lung and small intestine. In contrast, the disaccharide compositions of chondroitin sulfate were not primarily altered, indicating specificity of Sulfs for heparan sulfate. For Sulf1, but not for Sulf2, mRNA expression levels in eight organs of wild-type mice were highly correlated with increases in ΔUA2S-GlcNS6S in the corresponding organs of knock-out mice. Moreover, overall changes in heparan sulfate compositions were greater in Sulf1(-/-) mice than in Sulf2(-/-) mice despite lower levels of Sulf1 mRNA expression, suggesting predominant roles of Sulf1 in heparan sulfate desulfation and distinct regulation of Sulf activities in vivo. Sulf1 and Sulf2 mRNAs were differentially expressed in restricted types of cells in organs, and consequently, the sulfation patterns of heparan sulfate were locally and distinctly altered in Sulf1 and Sulf2 knock-out mice. These findings indicate that Sulf1 and Sulf2 differentially contribute to the generation of organ-specific sulfation patterns of heparan sulfate.


Asunto(s)
Espacio Extracelular/enzimología , Heparitina Sulfato/metabolismo , Proteínas/metabolismo , Sulfotransferasas/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Espacio Extracelular/genética , Heparitina Sulfato/química , Riñón/enzimología , Riñón/metabolismo , Pulmón/enzimología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estructura Molecular , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Especificidad de Órganos , Proteínas/genética , Sulfotransferasas/genética
8.
Nat Genet ; 36(1): 83-7, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14702043

RESUMEN

The biliary system, pancreas and liver all develop from the nearby foregut at almost the same time in mammals. The molecular mechanisms that determine the identity of each organ in this complex area are unknown. Hes1 encodes the basic helix-loop-helix protein Hes1 (ref. 1), which represses positive basic helix-loop-helix genes such as Neurog3 (ref. 3). Expression of Hes1 is controlled by the evolutionarily conserved Notch pathway. Hes1 operates as a general negative regulator of endodermal endocrine differentiation, and defects in Notch signaling lead to accelerated pancreatic endocrine differentiation. Mutations in JAG1, encoding a Notch ligand, cause the Alagille syndrome in humans, characterized by poor development of the biliary system, suggesting that the Notch pathway is also involved in normal biliary development. Here we show that Hes1 is expressed in the extrahepatic biliary epithelium throughout development and that Hes1-deficient mice have gallbladder agenesis and severe hypoplasia of extrahepatic bile ducts. Biliary epithelium in Hes1-/- mice ectopically expresses the proendocrine gene Neurog3 (refs. 12,13), differentiates into endocrine and exocrine cells and forms acini and islet-like structures in the mutant bile ducts. Thus, biliary epithelium has the potential for pancreatic differentiation and Hes1 determines biliary organogenesis by preventing the pancreatic differentiation program, probably by directly repressing transcription of Neurog3.


Asunto(s)
Sistema Biliar/embriología , Páncreas/embriología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Vesícula Biliar/embriología , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Ratones , Ratones Mutantes , Morfogénesis , Proteínas del Tejido Nervioso/genética , Transducción de Señal , Factor de Transcripción HES-1 , Factores de Transcripción/genética
9.
Biol Open ; 12(11)2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37795611

RESUMEN

Autotaxin, encoded by the Enpp2 gene, is an exoenzyme that produces lysophosphatidic acid, thereby regulating many biologic functions. We previously reported that Enpp2 mRNA was abundantly expressed in yolk sac visceral endoderm (VE) cells and that Enpp2-/- mice were lethal at embryonic day 9.5 owing to angiogenic defects in the yolk sac. Enpp2-/- mice showed lysosome fragmentation in VE cells and embryonic abnormalities including allantois malformation, neural tube defects, no axial turning, and head cavity formation. However, whether the defects in endocytic vesicle formation affect membrane trafficking in VE cells remained to be directly examined. In this study, we found that pinocytosis, transcytosis, and secretion of angiogenic factors such as vascular endothelial growth factor and transforming growth factor ß1 were impaired in Enpp2-/- VE cells. Moreover, pharmacologic inhibition of membrane trafficking phenocopied the defects of Enpp2-/- mice. These findings demonstrate that Enpp2 promotes endocytosis and secretion of angiogenic factors in VE cells, thereby regulating angiogenesis/vasculogenesis and embryonic development.


Asunto(s)
Hidrolasas Diéster Fosfóricas , Saco Vitelino , Animales , Femenino , Ratones , Embarazo , Diferenciación Celular , Desarrollo Embrionario , Endodermo , Factor A de Crecimiento Endotelial Vascular , Saco Vitelino/irrigación sanguínea , Hidrolasas Diéster Fosfóricas/metabolismo
10.
Genes Cells ; 16(10): 1012-21, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21895889

RESUMEN

Rho-associated coiled-coil-forming protein serine/threonine kinase (ROCK) consisting of two isoforms, ROCK-I and ROCK-II, functions downstream of the small GTPase Rho for assembly of actomyosin bundles. To examine the role of ROCK isoforms in vivo, we previously generated and examined mice deficient in each of the two isoforms individually. Here, we further examined the in vivo role of ROCK isoforms by generating mice deficient in both isoforms. Cross-mating of ROCK-I(+/-) ROCK-II(+/-) double heterozygous mice showed that all of the ROCK-I(-/-) ROCK-II(-/-) homozygous mice die in utero before 9.5 days post-coitum (dpc) and ROCK-I(-/-) ROCK-II(+/-) homo-heterozygous or ROCK-I(+/-) ROCK-II(-/-) hetero-homozygous mice die during a period from 9.5 to 12.5 dpc, whereas mice of other genotypes survive until 12.5 dpc with the expected Mendelian ratio. All of the ROCK-I(+/-) ROCK-II(-/-) or ROCK-I(-/-) ROCK-II(+/-) mice showed impaired body turning and defective vascular remodeling in the yolk sac. Impairment of vascular remodeling was also observed in wild-type embryos treated ex vivo with a ROCK inhibitor, Y-27632. These results suggest that ROCK isoforms function redundantly during embryogenesis and play a critical role in vascular development.


Asunto(s)
Saco Vitelino/irrigación sanguínea , Saco Vitelino/enzimología , Quinasas Asociadas a rho/deficiencia , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis/genética , Mutación/genética , Neovascularización Patológica/enzimología , Neovascularización Patológica/genética , Fenotipo , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
11.
Dev Dyn ; 240(2): 413-21, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21246658

RESUMEN

Autotaxin (ATX) is a lysophospholipid-generating exoenzyme expressed in embryonic and adult neural tissues. We previously showed that ATX is expressed in the neural organizing centers, anterior head process, and midbrain-hindbrain boundary (MHB). To elucidate the role of ATX during neural development, here we examined the neural phenotypes of ATX-deficient mice. Expression analysis of neural marker genes revealed that lateral expansion of the rostral forebrain is reduced and establishment of the MHB is compromised as early as the late headfold stage in ATX mutant embryos. Moreover, ATX mutant embryos fail to complete cranial neural tube closure. These results indicate that ATX is essential for cranial neurulation and MHB establishment.


Asunto(s)
Mesencéfalo/embriología , Complejos Multienzimáticos/metabolismo , Neurulación/fisiología , Fosfodiesterasa I/metabolismo , Pirofosfatasas/metabolismo , Rombencéfalo/embriología , Animales , Biomarcadores/metabolismo , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Hibridación in Situ , Mesencéfalo/anatomía & histología , Ratones , Ratones Noqueados , Complejos Multienzimáticos/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción Otx/genética , Factores de Transcripción Otx/metabolismo , Fosfodiesterasa I/genética , Hidrolasas Diéster Fosfóricas , Pirofosfatasas/genética , Rombencéfalo/anatomía & histología , Proteína Homeobox SIX3
12.
Magn Reson Imaging ; 88: 123-131, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35131262

RESUMEN

During brain development, neural circuits are formed through cellular differentiation, cell migration, axon guidance, and synaptogenic processes by the coordinated actions of many genes. Abnormalities in neural development, especially connectivity defects, can result in psychiatric disorders, such as schizophrenia and autism. Recent advances in diffusion tensor imaging have enabled us to examine the brain's macroscopic nerve trajectories. In this study, we investigated the abnormalities of the commissural fibers that connect the left and right cerebral hemispheres in mice lacking heparan sulfate 6-O endosulfatases, Sulf1 and Sulf2 (Sulf1/2), which are extracellular enzymes that remove 6-O sulfate from heparan sulfate and thereby modulate the function of axon guidance factors. We previously demonstrated that Sulf1/2 double knockout (DKO) mouse embryos harbored defects in their corticospinal tract and that some of these DKO mice experienced corpus callosum agenesis. However, abnormalities of the commissural fibers in the adult DKO brain have not been systematically assessed. In this study, we investigated commissural fiber abnormalities in these mice by the combined use of radiological and histological analyses. First, we acquired diffusion-weighted images and three-dimensional-T2 weighted images of adult brains using a 9.4 T animal magnetic resonance imaging system and found that Sulf1/2 DKO mice had a smaller corpus callosum and dorsal hippocampal commissure. Next, we performed myelin staining and anterograde tracing, revealing that the dorsal hippocampal commissure was elongated in a rostral direction. These results suggest that Sulf1/2 play an important role in the formation of commissural tracts and that diffusion tensor imaging associated with microscopic analysis is a powerful tool to clarify nerve tract abnormalities.


Asunto(s)
Imagen de Difusión Tensora , Sulfotransferasas , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Heparitina Sulfato , Humanos , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sulfotransferasas/genética , Sulfotransferasas/metabolismo
13.
J Cell Biol ; 174(6): 773-8, 2006 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-16966419

RESUMEN

Specific sulfation sequence of heparan sulfate (HS) contributes to the selective interaction between HS and various proteins in vitro. To clarify the in vivo importance of HS fine structures, we characterized the functions of the Drosophila HS 2-O and 6-O sulfotransferase (Hs2st and Hs6st) genes in FGF-mediated tracheal formation. We found that mutations in Hs2st or Hs6st had unexpectedly little effect on tracheal morphogenesis. Structural analysis of mutant HS revealed not only a loss of corresponding sulfation, but also a compensatory increase of sulfation at other positions, which maintains the level of HS total charge. The restricted phenotypes of Hsst mutants are ascribed to this compensation because FGF signaling is strongly disrupted by Hs2st; Hs6st double mutation, or by overexpression of 6-O sulfatase, an extracellular enzyme which removes 6-O sulfate groups without increasing 2-O sulfation. These findings suggest that the overall sulfation level is more important than strictly defined HS fine structures for FGF signaling in some developmental contexts.


Asunto(s)
Drosophila melanogaster/embriología , Factores de Crecimiento de Fibroblastos/metabolismo , Heparitina Sulfato/metabolismo , Sulfotransferasas/metabolismo , Tráquea/embriología , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Heparitina Sulfato/química , Masculino , Estructura Molecular , Mutación/genética , Organogénesis/genética , Transducción de Señal/fisiología , Sulfotransferasas/genética , Ésteres del Ácido Sulfúrico/metabolismo , Tráquea/citología , Tráquea/metabolismo
14.
Artículo en Inglés | MEDLINE | ID: mdl-21795788

RESUMEN

Coiled-coil DIX1 (Ccd1) is a positive regulator that activates the canonical Wnt signalling pathway by inhibiting the degradation of the key signal transducer ß-catenin. The C-terminal DIX domain of Ccd1 plays an important role in the regulation of signal transduction through homo-oligomerization and protein complex formation with other DIX domain-containing proteins, i.e. axin and dishevelled proteins. Here, the expression, purification, crystallization and X-ray data collection of the Ccd1 DIX domain are reported. The crystals of the Ccd1 DIX domain belonged to space group P2(1)2(1)2(1), with unit-cell parameters a=72.9, b=75.7, c=125.6 Å. An X-ray diffraction data set was collected at 3.0 Šresolution.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/química , Transducción de Señal , Animales , Cristalografía por Rayos X , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Proteínas Wnt/metabolismo
15.
Front Neuroanat ; 15: 726718, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34489650

RESUMEN

The heparan sulfate 6-O-endosulfatases, Sulfatase 1 (Sulf1), and Sulfatase 2 (Sulf2), are extracellular enzymes that regulate cellular signaling by removing 6-O-sulfate from the heparan sulfate chain. Although previous studies have revealed that Sulfs are essential for normal development, their functions in the adult brain remain largely unknown. To gain insight into their neural functions, we used in situ hybridization to systematically examine Sulf1/2 mRNA expression in the adult mouse brain. Sulf1 and Sulf2 mRNAs showed distinct expression patterns, which is in contrast to their overlapping expression in the embryonic brain. In addition, we found that Sulf1 was distinctly expressed in the nucleus accumbens shell, the posterior tail of the striatum, layer 6 of the cerebral cortex, and the paraventricular nucleus of the thalamus, all of which are target areas of dopaminergic projections. Using double-labeling techniques, we showed that Sulf1-expressing cells in the above regions coincided with cells expressing the dopamine D1 and/or D2 receptor. These findings implicate possible roles of Sulf1 in modulation of dopaminergic transmission and dopamine-mediated behaviors.

16.
J Neurosci ; 29(13): 4312-21, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19339625

RESUMEN

The transcriptional corepressor SnoN is a critical regulator of axonal morphogenesis, but how SnoN drives axonal growth is unknown. Here, we report that gene-profiling analyses in cerebellar granule neurons reveal that the large majority of genes altered upon SnoN knockdown are surprisingly downregulated, suggesting that SnoN may activate transcription in neurons. Accordingly, we find that the transcriptional coactivator p300 interacts with SnoN, and p300 plays a critical role in SnoN-induced axon growth. We also identify the gene encoding the signaling scaffold protein Ccd1 as a critical target of SnoN in neurons. Ccd1 localizes to the actin cytoskeleton, is enriched at axon terminals in neurons, and activates the axon growth-promoting kinase JNK (c-Jun N-terminal protein kinase). Knockdown of Ccd1 in neurons reduces axonal length and suppresses the ability of SnoN to promote axonal growth. Importantly, Ccd1 knockdown in rat pups profoundly impairs the formation of granule neuron parallel fiber axons in the rat cerebellar cortex in vivo. These findings define a novel SnoN-Ccd1 link that promotes axonal growth in the mammalian brain, with important implications for axonal development and regeneration.


Asunto(s)
Axones/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Morfogénesis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Células Cultivadas , Cerebelo/citología , Chlorocebus aethiops , Proteína p300 Asociada a E1A/genética , Proteína p300 Asociada a E1A/metabolismo , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , MAP Quinasa Quinasa 4/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Análisis por Micromatrices/métodos , Morfogénesis/genética , Proteínas del Tejido Nervioso/genética , Interferencia de ARN/fisiología , ARN Mensajero/metabolismo , Ratas , Ratas Long-Evans , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética , Transfección/métodos , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno
17.
J Biol Chem ; 284(48): 33561-70, 2009 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-19808661

RESUMEN

Autotaxin, a lysophospholipase D encoded by the Enpp2 gene, is an exoenzyme that produces lysophosphatidic acid in the extracellular space. Lysophosphatidic acid acts on specific G protein-coupled receptors, thereby regulating cell growth, migration, and survival. Previous studies have revealed that Enpp2(-/-) mouse embryos die at about embryonic day (E) 9.5 because of angiogenic defects in the yolk sac. However, what cellular defects occur in Enpp2(-/-) embryos and what intracellular signaling pathways are involved in the phenotype manifestation remain unknown. Here, we show that Enpp2 is required to form distinctive large lysosomes in the yolk sac visceral endoderm cells. From E7.5 to E9.5, Enpp2 mRNA is abundantly expressed in the visceral endoderm cells. In Enpp2(-/-) mouse embryos, lysosomes in the visceral endoderm cells are fragmented. By using a whole embryo culture system combined with specific pharmacological inhibitors for intracellular signaling molecules, we show that lysophosphatidic acid receptors and the Rho-Rho-associated coiled-coil containing protein kinase (ROCK)-LIM kinase pathway are required to form large lysosomes. In addition, electroporation of dominant negative forms of Rho, ROCK, or LIM kinase also leads to the size reduction of lysosomes in wild-type visceral endoderm cells. In Enpp2(-/-) visceral endoderm cells, the steady-state levels of cofilin phosphorylation and actin polymerization are reduced. In addition, perturbations of actin turnover dynamics by actin inhibitors cytochalasin B and jasplakinolide result in the defect in lysosome formation. These results suggest that constitutive activation of the Rho-ROCK-LIM kinase pathway by extracellular production of lysophosphatidic acid by the action of autotaxin is required to maintain the large size of lysosomes in visceral endoderm cells.


Asunto(s)
Lisofosfolípidos/metabolismo , Lisosomas/metabolismo , Complejos Multienzimáticos/metabolismo , Fosfodiesterasa I/metabolismo , Pirofosfatasas/metabolismo , Saco Vitelino/metabolismo , Factores Despolimerizantes de la Actina/genética , Factores Despolimerizantes de la Actina/metabolismo , Animales , Endodermo/citología , Endodermo/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hibridación in Situ , Quinasas Lim/genética , Quinasas Lim/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Microscopía Confocal , Microscopía Electrónica , Complejos Multienzimáticos/genética , Fosfodiesterasa I/genética , Hidrolasas Diéster Fosfóricas , Fosforilación , Pirofosfatasas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Saco Vitelino/citología , Saco Vitelino/ultraestructura , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoB/genética , Proteína de Unión al GTP rhoB/metabolismo
18.
Biochem Biophys Res Commun ; 400(1): 66-71, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20692235

RESUMEN

Autotaxin, encoded by the Enpp2 gene, generates lysophosphatidic acid (LPA) extracellularly, eliciting various cellular responses through specific LPA receptors. Previous studies have revealed that Enpp2(-/-) mice die at E9.5 owing to angiogenic defects in the yolk sac. Moreover, Enpp2(-/-) embryos show growth retardation, allantois malformation, no axial turning, and head cavity formation. We have also demonstrated that lysosome biogenesis is impaired in yolk sac visceral endoderm cells of Enpp2(-/-) embryos as a result of the downregulation of the Rho-ROCK (Rho-associated coiled-coil containing protein kinase)-LIM kinase pathway. In this study, we examine what signaling defect(s) is responsible for head cavity formation and yolk sac angiogenic defects. By using a whole embryo culture system, we show that 10 µM Ki16425, an antagonist for the LPA receptors, induces head cavity formation and yolk sac angiogenic defects in wild-type embryos. Moreover, 1 µM Ki16425 induces both phenotypes in Enpp2 heterozygous embryos at significantly higher incidence than in wild-type embryos, suggesting an interaction between autotaxin and LPA receptor signaling. Furthermore, we show that inhibition of the Rho-ROCK pathway induces head cavity formation, whereas multiple pathways are involved in yolk sac angiogenic defects. These results reveal the signal transduction defects that underlie the abnormalities in Enpp2(-/-) embryos.


Asunto(s)
Embrión de Mamíferos/anomalías , Cabeza/anomalías , Complejos Multienzimáticos/genética , Fosfodiesterasa I/genética , Hidrolasas Diéster Fosfóricas/genética , Pirofosfatasas/genética , Receptores del Ácido Lisofosfatídico/metabolismo , Factor Rho/metabolismo , Quinasas Asociadas a rho/metabolismo , Actinas/metabolismo , Animales , Vasos Sanguíneos/anomalías , Embrión de Mamíferos/metabolismo , Ratones , Ratones Mutantes , Neovascularización Fisiológica/genética , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Factor Rho/antagonistas & inhibidores , Transducción de Señal/genética , Saco Vitelino/anomalías , Saco Vitelino/irrigación sanguínea , Quinasas Asociadas a rho/antagonistas & inhibidores
19.
Biochem Biophys Res Commun ; 391(1): 107-12, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19900405

RESUMEN

Heparan sulfate 6-O-endosufatases Sulf1 and Sulf2 hydrolyze the 6-O-sulfate of the glucosamine residues in heparin and heparan sulfate, thereby regulating multiple signaling pathways. A previous study reported that human Sulf1 and Sulf2 were proteolytically processed in a manner sensitive to a furin inhibitor. However, the exact sites of cleavage, the sequence motifs for proteolysis, and the effect of the cleavage on enzyme activity remain unknown. Here we show that the cleavage of rat Sulf2 (also called SulfFP2) occurs at two arginine residues, 543 and 570, in the hydrophilic domain. Both sites reside in the consensus sequence for the cleavage by furin-type proprotein convertases, and the consensus motifs are essential for cleavages. The cleavage at arginine 570 is sensitive to a furin inhibitor. Furthermore, the uncleavable form of SulfFP2 shows sulfatase activity comparable to the cleavable SulfFP2, indicating that the cleavage is not indispensable for activation of SulfFP2.


Asunto(s)
Arginina/metabolismo , Furina/metabolismo , Sulfotransferasas/metabolismo , Secuencia de Aminoácidos , Animales , Arginina/química , Arginina/genética , Furina/química , Humanos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Ratas , Sulfotransferasas/genética
20.
Data Brief ; 23: 103718, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31372387

RESUMEN

The corticospinal tract (CST) has a complex and long trajectory throughout the brain. Semaphorin 6A (Sema6A), a member of the semaphorin family, is one of the important regulators of CST axon guidance. Previous studies have shown that Sema6A knockout (KO) mice have CST defects at the midbrain-hindbrain boundary and medulla [1]. However, the route of the aberrant fibers remained unknown. Therefore here, to track the trajectory of the abnormal fibers, 3D images of the CST in adult mice were reconstructed from serial brain sections stained with anti-PKCγ antibody. Sema6A mutant brains showed CST defects that were more complex and variable than previously thought. In addition, 3D analysis helped us to identify a few new patterns of abnormal fibers. For more information about the data, please refer to an original research article, which has been recently published by Brain Research, "Remarkable complexity and variability of corticospinal tract defects in adult Semaphorin 6A knockout mice" [2].

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA