Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Glycobiology ; 31(11): 1531-1542, 2021 12 18.
Article in English | MEDLINE | ID: mdl-34324645

ABSTRACT

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.


Subject(s)
Heparitin Sulfate/metabolism , Lateral Ventricles/metabolism , Sulfatases/metabolism , Sulfotransferases/metabolism , Animals , Extracellular Matrix , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Neurogenesis , Stem Cell Niche , Sulfatases/deficiency , Sulfotransferases/deficiency
2.
Dev Neurosci ; 39(5): 361-374, 2017.
Article in English | MEDLINE | ID: mdl-28490013

ABSTRACT

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.


Subject(s)
Hedgehog Proteins/metabolism , Motor Neurons/metabolism , Oligodendrocyte Precursor Cells/metabolism , Signal Transduction , Sulfatases/metabolism , Sulfotransferases/metabolism , Animals , Cell Differentiation/physiology , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology , Spinal Cord/metabolism , Sulfatases/genetics , Sulfotransferases/genetics
3.
Am J Physiol Renal Physiol ; 310(5): F395-408, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26764203

ABSTRACT

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.


Subject(s)
Diabetic Nephropathies/enzymology , Heparitin Sulfate/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Kidney Glomerulus/drug effects , Receptors, Growth Factor/agonists , Signal Transduction/drug effects , Sulfatases/metabolism , Sulfotransferases/metabolism , Animals , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Genetic Predisposition to Disease , Kidney Glomerulus/enzymology , Kidney Glomerulus/pathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-sis/pharmacology , Receptors, Growth Factor/metabolism , Smad Proteins, Receptor-Regulated/metabolism , Sulfatases/deficiency , Sulfatases/genetics , Sulfotransferases/deficiency , Sulfotransferases/genetics , Time Factors , Tissue Culture Techniques , Transforming Growth Factor beta/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
4.
Dev Dyn ; 244(2): 168-80, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25370455

ABSTRACT

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.


Subject(s)
Avian Proteins/biosynthesis , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Organ of Corti/embryology , Sulfatases/biosynthesis , Sulfotransferases/biosynthesis , Animals , Chick Embryo , Mice , Organ of Corti/cytology , Signal Transduction/physiology
5.
MicroPubl Biol ; 20242024.
Article in English | MEDLINE | ID: mdl-38725939

ABSTRACT

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.

6.
J Biol Chem ; 287(12): 9579-90, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22298771

ABSTRACT

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.


Subject(s)
Extracellular Space/enzymology , Heparitin Sulfate/metabolism , Proteins/metabolism , Sulfotransferases/metabolism , Animals , Brain/enzymology , Brain/metabolism , Extracellular Space/genetics , Heparitin Sulfate/chemistry , Kidney/enzymology , Kidney/metabolism , Lung/enzymology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Organ Specificity , Proteins/genetics , Sulfotransferases/genetics
7.
Nat Genet ; 36(1): 83-7, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14702043

ABSTRACT

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.


Subject(s)
Biliary Tract/embryology , Pancreas/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , Gallbladder/embryology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Mice , Mice, Mutant Strains , Morphogenesis , Nerve Tissue Proteins/genetics , Signal Transduction , Transcription Factor HES-1 , Transcription Factors/genetics
8.
Biol Open ; 12(11)2023 11 15.
Article in English | MEDLINE | ID: mdl-37795611

ABSTRACT

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.


Subject(s)
Phosphoric Diester Hydrolases , Yolk Sac , Animals , Female , Mice , Pregnancy , Cell Differentiation , Embryonic Development , Endoderm , Vascular Endothelial Growth Factor A , Yolk Sac/blood supply , Phosphoric Diester Hydrolases/metabolism
9.
Dev Dyn ; 240(2): 413-21, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21246658

ABSTRACT

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.


Subject(s)
Mesencephalon/embryology , Multienzyme Complexes/metabolism , Neurulation/physiology , Phosphodiesterase I/metabolism , Pyrophosphatases/metabolism , Rhombencephalon/embryology , Animals , Biomarkers/metabolism , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Eye Proteins/genetics , Eye Proteins/metabolism , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization , Mesencephalon/anatomy & histology , Mice , Mice, Knockout , Multienzyme Complexes/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism , Phosphodiesterase I/genetics , Phosphoric Diester Hydrolases , Pyrophosphatases/genetics , Rhombencephalon/anatomy & histology , Homeobox Protein SIX3
10.
Magn Reson Imaging ; 88: 123-131, 2022 05.
Article in English | MEDLINE | ID: mdl-35131262

ABSTRACT

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.


Subject(s)
Diffusion Tensor Imaging , Sulfotransferases , Animals , Brain/diagnostic imaging , Brain/metabolism , Heparitin Sulfate , Humans , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Mice, Knockout , Sulfotransferases/genetics , Sulfotransferases/metabolism
11.
Nat Commun ; 13(1): 4039, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35864121

ABSTRACT

The dorsal raphe nucleus (DRN) is known to control aggressive behavior in mice. Here, we found that glutamatergic projections from the lateral habenula (LHb) to the DRN were activated in male mice that experienced pre-exposure to a rival male mouse ("social instigation") resulting in heightened intermale aggression. Both chemogenetic and optogenetic suppression of the LHb-DRN projection blocked heightened aggression after social instigation in male mice. In contrast, inhibition of this pathway did not affect basal levels of aggressive behavior, suggesting that the activity of the LHb-DRN projection is not necessary for the expression of species-typical aggressive behavior, but required for the increase of aggressive behavior resulting from social instigation. Anatomical analysis showed that LHb neurons synapse on non-serotonergic DRN neurons that project to the ventral tegmental area (VTA), and optogenetic activation of the DRN-VTA projection increased aggressive behaviors. Our results demonstrate that the LHb glutamatergic inputs to the DRN promote aggressive arousal induced by social instigation, which contributes to aggressive behavior by activating VTA-projecting non-serotonergic DRN neurons as one of its potential targets.


Subject(s)
Dorsal Raphe Nucleus , Habenula , Aggression/physiology , Animals , Arousal , Dorsal Raphe Nucleus/physiology , Habenula/physiology , Male , Mice , Neural Pathways/physiology , Neurons/metabolism
12.
Article in English | MEDLINE | ID: mdl-21795788

ABSTRACT

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.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Signal Transduction , Animals , Crystallography, X-Ray , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Wnt Proteins/metabolism
13.
Front Neuroanat ; 15: 726718, 2021.
Article in English | MEDLINE | ID: mdl-34489650

ABSTRACT

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.

14.
J Biol Chem ; 284(48): 33561-70, 2009 Nov 27.
Article in English | MEDLINE | ID: mdl-19808661

ABSTRACT

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.


Subject(s)
Lysophospholipids/metabolism , Lysosomes/metabolism , Multienzyme Complexes/metabolism , Phosphodiesterase I/metabolism , Pyrophosphatases/metabolism , Yolk Sac/metabolism , Actin Depolymerizing Factors/genetics , Actin Depolymerizing Factors/metabolism , Animals , Endoderm/cytology , Endoderm/metabolism , Female , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Situ Hybridization , Lim Kinases/genetics , Lim Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron , Multienzyme Complexes/genetics , Phosphodiesterase I/genetics , Phosphoric Diester Hydrolases , Phosphorylation , Pyrophosphatases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Yolk Sac/cytology , Yolk Sac/ultrastructure , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , rhoB GTP-Binding Protein/genetics , rhoB GTP-Binding Protein/metabolism
15.
Biochem Biophys Res Commun ; 400(1): 66-71, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20692235

ABSTRACT

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.


Subject(s)
Embryo, Mammalian/abnormalities , Head/abnormalities , Multienzyme Complexes/genetics , Phosphodiesterase I/genetics , Phosphoric Diester Hydrolases/genetics , Pyrophosphatases/genetics , Receptors, Lysophosphatidic Acid/metabolism , Rho Factor/metabolism , rho-Associated Kinases/metabolism , Actins/metabolism , Animals , Blood Vessels/abnormalities , Embryo, Mammalian/metabolism , Mice , Mice, Mutant Strains , Neovascularization, Physiologic/genetics , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Rho Factor/antagonists & inhibitors , Signal Transduction/genetics , Yolk Sac/abnormalities , Yolk Sac/blood supply , rho-Associated Kinases/antagonists & inhibitors
16.
Biochem Biophys Res Commun ; 391(1): 107-12, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19900405

ABSTRACT

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.


Subject(s)
Arginine/metabolism , Furin/metabolism , Sulfotransferases/metabolism , Amino Acid Sequence , Animals , Arginine/chemistry , Arginine/genetics , Furin/chemistry , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Rats , Sulfotransferases/genetics
17.
Brain Res ; 1710: 209-219, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30599138

ABSTRACT

The corticospinal tract (CST) has a complex and long trajectory that originates in the cerebral cortex and ends in the spinal cord. Semaphorin 6A (Sema6A), a member of the semaphorin family, is an important regulator of CST axon guidance. Previous studies have shown that postnatal Sema6A mutant mice have CST defects at the midbrain-hindbrain boundary and medulla. However, the routes the aberrant fibers take throughout the Sema6A mutant brain remain unknown. In this study, we performed 3D reconstruction of immunostained CST fibers to reevaluate the details of the abnormal CST trajectories in the brains of adult Sema6A mutant mice. Our results showed that the axon guidance defects reported in early postnatal mutants were consistently observed in adulthood. Those abnormal trajectories revealed by 3D analysis of brain sections were, however, more complex and variable than previously thought. In addition, 3D analysis allowed us to identify a few new patterns of aberrant projections. First, a subset of fibers that separated from and descended in parallel to the main bundle projected laterally at the caudal pons, subsequently changed direction by turning caudally, and extended to the medulla. Second, some abnormal fibers returned to the correct trajectory after deviating substantially from the original tract. Third, some fibers reached the pyramidal decussation normally but did not enter the dorsal funiculus. Section immunostaining combined with 3D reconstruction is a powerful method to track long projection fibers and to examine the entire nerve tracts of both normal and abnormal animals.


Subject(s)
Brain/growth & development , Pyramidal Tracts/growth & development , Semaphorins/physiology , Animals , Brain/cytology , Mice, Knockout , Neuroanatomical Tract-Tracing Techniques , Pyramidal Tracts/cytology , Semaphorins/genetics
18.
Data Brief ; 23: 103718, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31372387

ABSTRACT

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].

19.
Front Mol Neurosci ; 12: 333, 2019.
Article in English | MEDLINE | ID: mdl-32038163

ABSTRACT

The corticospinal tract (CST) plays an important role in controlling voluntary movement. Because the CST has a long trajectory throughout the brain toward the spinal cord, many axon guidance molecules are required to navigate the axons correctly during development. Previously, we found that double-knockout (DKO) mouse embryos lacking the heparan sulfate endosulfatases, Sulf1 and Sulf2, showed axon guidance defects of the CST owing to the abnormal accumulation of Slit2 protein on the brain surface. However, postnatal development of the CST, especially the pyramidal decussation and spinal cord projection, could not be assessed because DKO mice on a C57BL/6 background died soon after birth. We recently found that Sulf1/2 DKO mice on a mixed C57BL/6 and CD-1/ICR background can survive into adulthood and therefore investigated the anatomy and function of the CST in the adult DKO mice. In Sulf1/2 DKO mice, abnormal dorsal deviation of the CST fibers on the midbrain surface persisted after maturation of the CST. At the pyramidal decussation, some CST fibers located near the midline crossed the midline, whereas others located more laterally extended ipsilaterally. In the spinal cord, the crossed CST fibers descended in the dorsal funiculus on the contralateral side and entered the contralateral gray matter normally, whereas the uncrossed fibers descended in the lateral funiculus on the ipsilateral side and entered the ipsilateral gray matter. As a result, the CST fibers that originated from 1 side of the brain projected bilaterally in the DKO spinal cord. Consistently, microstimulation of 1 side of the motor cortex evoked electromyogram responses only in the contralateral forelimb muscles of the wild-type mice, whereas the same stimulation evoked bilateral responses in the DKO mice. The functional consequences of the CST defects in the Sulf1/2 DKO mice were examined using the grid-walking, staircase, and single pellet-reaching tests, which have been used to evaluate motor function in mice. Compared with the wild-type mice, the Sulf1/2 DKO mice showed impaired performance in these tests, indicating deficits in motor function. These findings suggest that disruption of Sulf1/2 genes leads to both anatomical and functional defects of the CST.

20.
Curr Biol ; 13(1): 73-7, 2003 Jan 08.
Article in English | MEDLINE | ID: mdl-12526749

ABSTRACT

Wnt signaling plays a crucial role in directing cell differentiation, polarity, and growth. In the canonical pathway, Wnt receptors activate Dishevelled (Dvl), which then blocks the degradation of a key signal transducer, beta-catenin, leading to the nuclear accumulation of beta-catenin and induction of Wnt target genes through TCF/LEF family transcription factors. Here we identified a novel zebrafish gene encoding Ccd1, which possesses a DIX (Dishevelled-Axin) domain. DIX domains are essential for the signal transduction of two major Wnt downstream mediators, Dvl and Axin. Ccd1 formed homomeric and heteromeric complexes with Dvl and Axin and activated TCF-dependent transcription in vitro. In addition, overexpression of ccd1 in zebrafish embryos led to a reduction in the size of the eyes and forebrain (posteriorization), as seen with wnt8 overexpression, whereas a dominant-negative ccd1 (DN-ccd1) caused the opposite phenotype. Furthermore, the Wnt activation phenotype induced by ccd1 was inhibited by the expression of axin1 or DN-ccd1, and the wnt8 overexpression phenotype was rescued by DN-ccd1, suggesting that Ccd1 functions downstream of the Wnt receptor and upstream of Axin. These results indicate that Ccd1 is a novel positive regulator in this Wnt signaling pathway during zebrafish neural patterning.


Subject(s)
Body Patterning/physiology , Nervous System/embryology , Proteins/metabolism , Repressor Proteins , Signal Transduction , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish/embryology , Amino Acid Sequence , Animals , Axin Protein , Binding Sites , Cytoskeletal Proteins , DNA-Binding Proteins/metabolism , Embryo, Nonmammalian , Eye Abnormalities/genetics , Eye Proteins , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Lymphoid Enhancer-Binding Factor 1 , Molecular Sequence Data , Nervous System/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Sequence Homology, Amino Acid , Transcription Factors/metabolism , Transfection , Wnt Proteins , Zebrafish/genetics
SELECTION OF CITATIONS
SEARCH DETAIL