Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Biochem J ; 473(5): 661-72, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26699903

RESUMEN

SCUBE1 (S1), a secreted and membrane-bound glycoprotein, has a modular protein structure composed of an N-terminal signal peptide sequence followed by nine epidermal growth factor (EGF)-like repeats, a spacer region and three cysteine-rich (CR) motifs with multiple potential N-linked glycosylation sites, and one CUB domain at the C-terminus. Soluble S1 is a biomarker of platelet activation but an active participant of thrombosis via its adhesive EGF-like repeats, whereas its membrane-associated form acts as a bone morphogenetic protein (BMP) co-receptor in promoting BMP signal activity. However, the mechanism responsible for the membrane tethering and the biological importance of N-glycosylation of S1 remain largely unknown. In the present study, molecular mapping analysis identified a polycationic segment (amino acids 501-550) in the spacer region required for its membrane tethering via electrostatic interactions possibly with the anionic heparan sulfate proteoglycans. Furthermore, deglycosylation by peptide N-glycosidase F treatment revealed that N-glycans within the CR motif are essential for membrane recruitment through lectin-mediated surface retention. Injection of mRNA encoding zebrafish wild-type but not N-glycan-deficient scube1 restores the expression of haematopoietic and erythroid markers (scl and gata1) in scube1-knockdown embryos. We describe novel mechanisms in targeting S1 to the plasma membrane and demonstrate that N-glycans are required for S1 functions during primitive haematopoiesis in zebrafish.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de la Membrana/metabolismo , Oligosacáridos/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Proteínas de Unión al Calcio , Membrana Celular/metabolismo , Glicosilación , Células HEK293 , Hematopoyesis , Humanos , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Mutación , Unión Proteica , Estructura Terciaria de Proteína , Transducción de Señal , Electricidad Estática , Pez Cebra/sangre
2.
J Biol Chem ; 289(27): 18928-42, 2014 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-24849601

RESUMEN

SCUBE3 (signal peptide CUB-EGF-like domain-containing protein 3) belongs to a newly identified secreted and cell membrane-associated SCUBE family, which is evolutionarily conserved in vertebrates. Scube3 is predominantly expressed in a variety of developing tissues in mice such as somites, neural tubes, and limb buds. However, its function during development remains unclear. In this study, we first showed that knockdown of SCUBE3 in C2C12 myoblasts inhibited FGF receptor 4 expression and FGF signaling, thus resulting in reduced myogenic differentiation. Furthermore, knockdown of zebrafish scube3 by antisense morpholino oligonucleotides specifically suppressed the expression of the myogenic marker myod1 within the lateral fast muscle precursors, whereas its expression in the adaxial slow muscle precursors was largely unaffected. Consistent with these findings, immunofluorescent staining of fast but not slow muscle myosin was markedly decreased in scube3 morphants. Further genetic studies identified fgf8 as a key regulator in scube3-mediated fast muscle differentiation in zebrafish. Biochemical and molecular analysis showed that SCUBE3 acts as a FGF co-receptor to augment FGF8 signaling. Scube3 may be a critical upstream regulator of fast fiber myogenesis by modulating fgf8 signaling during zebrafish embryogenesis.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Glicoproteínas/metabolismo , Desarrollo de Músculos , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Proteínas de Pez Cebra/metabolismo , Animales , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glicoproteínas/deficiencia , Glicoproteínas/genética , Células HEK293 , Humanos , Ratones , Proteína MioD/metabolismo , Oligonucleótidos Antisentido/genética , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Factores de Tiempo , Pez Cebra/embriología , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
3.
Mol Biol Evol ; 31(10): 2722-34, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25063441

RESUMEN

MicroRNAs (miRNAs) are a class of endogenous small noncoding RNAs that regulate gene expression either by degrading target mRNAs or by suppressing protein translation. miRNAs have been found to be involved in many biological processes, such as development, differentiation, and growth. However, the evolution of miRNA regulatory functions and networks has not been well studied. In this study, we conducted a cross-species analysis to study the evolution of cardiac miRNAs and their regulatory functions and networks. We found that conserved cardiac miRNA target genes have maintained highly conserved cardiac functions. Additionally, most of cardiac miRNA target genes in human with annotations of cardiac functions evolved from the corresponding homologous targets, which are also involved in heart development-related functions. On the basis of these results, we investigated the functional evolution of cardiac miRNAs and presented a functional evolutionary map. From this map, we identified the evolutionary time at which the cardiac miRNAs became involved in heart development or function and found that the biological processes of heart development evolved earlier than those of heart functions, for example, heart contraction/relaxation or cardiac hypertrophy. Our study of the evolution of the cardiac miRNA regulatory networks revealed the emergence of new regulatory functional branches during evolution. Furthermore, we discovered that early evolved cardiac miRNA target genes tend to participate in the early stages of heart development. This study sheds light on the evolution of developmental features of genes regulated by cardiac miRNAs.


Asunto(s)
Corazón/fisiología , MicroARNs/metabolismo , Miocardio/metabolismo , Animales , Secuencia de Bases , Secuencia Conservada , Evolución Molecular , Redes Reguladoras de Genes , Humanos , MicroARNs/genética
4.
Development ; 139(23): 4439-48, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23095890

RESUMEN

Left-right (L-R) patterning is essential for proper organ morphogenesis and function. Calcium fluxes in dorsal forerunner cells (DFCs) are known to regulate the formation of Kupffer's vesicle (KV), a central organ for establishing L-R asymmetry in zebrafish. Here, we identify the lipid mediator lysophosphatidic acid (LPA) as a regulator of L-R asymmetry in zebrafish embryos. LPA is produced by Autotaxin (Atx), a secreted lysophospholipase D, and triggers various cellular responses through activation of specific G protein-coupled receptors (Lpar1-6). Knockdown of Atx or LPA receptor 3 (Lpar3) by morpholino oligonucleotides perturbed asymmetric gene expression in lateral plate mesoderm and disrupted organ L-R asymmetries, whereas overexpression of lpar3 partially rescued those defects in both atx and lpar3 morphants. Similar defects were observed in embryos treated with the Atx inhibitor HA130 and the Lpar1-3 inhibitor Ki16425. Knockdown of either Atx or Lpar3 impaired calcium fluxes in DFCs during mid-epiboly stage and compromised DFC cohesive migration, KV formation and ciliogenesis. Application of LPA to DFCs rescued the calcium signal and laterality defects in atx morphants. This LPA-dependent L-R asymmetry is mediated via Wnt signaling, as shown by the accumulation of ß-catenin in nuclei at the dorsal side of both atx and lpar3 morphants. Our results suggest a major role for the Atx/Lpar3 signaling axis in regulating KV formation, ciliogenesis and L-R asymmetry via a Wnt-dependent pathway.


Asunto(s)
Tipificación del Cuerpo/genética , Lisofosfolípidos/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Receptores Purinérgicos P2/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Señalización del Calcio , Núcleo Celular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Isoxazoles/farmacología , Morfogénesis , Morfolinos/genética , Morfolinos/farmacología , Hidrolasas Diéster Fosfóricas/genética , Propionatos/farmacología , Receptores del Ácido Lisofosfatídico/genética , Receptores Purinérgicos P2/genética , Vía de Señalización Wnt , Pez Cebra/genética , Proteínas de Pez Cebra/genética , beta Catenina/metabolismo
5.
J Biol Chem ; 288(7): 5017-26, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23271740

RESUMEN

scube1 (signal peptide-CUB (complement protein C1r/C1s, Uegf, and Bmp1)-EGF domain-containing protein 1), the founding member of a novel secreted and cell surface SCUBE protein family, is expressed predominantly in various developing tissues in mice. However, its function in primitive hematopoiesis remains unknown. In this study, we identified and characterized zebrafish scube1 and analyzed its function by injecting antisense morpholino-oligonucleotide into embryos. Whole-mount in situ hybridization revealed that zebrafish scube1 mRNA is maternally expressed and widely distributed during early embryonic development. Knockdown of scube1 by morpholino-oligonucleotide down-regulated the expression of marker genes associated with early primitive hematopoietic precursors (scl) and erythroid (gata1 and hbbe1), as well as early (pu.1) and late (mpo and l-plastin) myelomonocytic lineages. However, the expression of an early endothelial marker fli1a and vascular morphogenesis appeared normal in scube1 morphants. Overexpression of bone morphogenetic protein (bmp) rescued the expression of scl in the posterior lateral mesoderm during early primitive hematopoiesis in scube1 morphants. Biochemical and molecular analysis revealed that Scube1 could be a BMP co-receptor to augment BMP signaling. Our results suggest that scube1 is critical for and functions at the top of the regulatory hierarchy of primitive hematopoiesis by modulating BMP activity during zebrafish embryogenesis.


Asunto(s)
Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/fisiología , Factor de Crecimiento Epidérmico/metabolismo , Regulación de la Expresión Génica , Hematopoyesis/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Membrana Celular/metabolismo , ADN Complementario/metabolismo , Eritrocitos/metabolismo , Células HEK293 , Humanos , Hibridación in Situ , Modelos Genéticos , Datos de Secuencia Molecular , Oligonucleótidos/genética , Estructura Terciaria de Proteína , Transducción de Señal , Pez Cebra
6.
Stem Cells ; 29(11): 1763-73, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21915944

RESUMEN

Lysophosphatidic acid (LPA), an extracellular lipid mediator, exerts multiple bioactivities through activating G protein-coupled receptors. LPA receptor 3 (LPA(3)) is a member of the endothelial differentiation gene family, which regulates differentiation and development of the circulation system. However, the relationship among the LPA receptors (LPARs) and erythropoiesis is still not clear. In this study, we found that erythroblasts expressed both LPA(1) and LPA(3), and erythropoietic defects were observed in zLPA(3) antisense morpholino oligonucleotide-injected zebrafish embryos. In human model, our results showed that LPA enhanced the erythropoiesis in the cord blood-derived human hematopoietic stem cells (hHSCs) with erythropoietin (EPO) addition in the plasma-free culture. When hHSCs were treated with Ki16425, an antagonist of LPA(1) and LPA(3), erythropoietic process of hHSCs was also blocked, as detected by mRNA and protein expressions of CD71 and GlyA. In the knockdown study, we further demonstrated that specific knockdown of LPA(3), not LPA(1), blocked the erythropoiesis. The translocation of ß-catenin into the nucleus, a downstream response of LPAR activation, was blocked by Ki16425 treatment. In addition, upregulation of erythropoiesis by LPA was also blocked by quercetin, an inhibitor of the ß-catenin/T-cell factor pathway. Furthermore, the enhancement of LPA on erythropoiesis was diminished by blocking c-Jun-activated kinase/signal transducer and activator of transcription and phosphatidylinositol 3-kinase/AKT activation, the downstream signaling pathways of EPO receptor, suggested that LPA might play a synergistic role with EPO to regulate erythropoietic process. In conclusion, we first reported that LPA participates in EPO-dependent erythropoiesis through activating LPA(3).


Asunto(s)
Eritropoyesis/efectos de los fármacos , Lisofosfolípidos/farmacología , Receptores del Ácido Lisofosfatídico/agonistas , Receptores del Ácido Lisofosfatídico/metabolismo , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Células Cultivadas , Embrión no Mamífero , Citometría de Flujo , Glicoproteínas/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Isoxazoles/farmacología , Péptidos/metabolismo , Propionatos/farmacología , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Pez Cebra
7.
Cardiovasc Res ; 118(4): 1074-1087, 2022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33788916

RESUMEN

AIMS: The secreted and membrane-anchored signal peptide-CUB-EGF domain-containing proteins (SCUBE) gene family composed of three members was originally identified from endothelial cells (ECs). We recently showed that membrane SCUBE2 binds vascular endothelial growth factor (VEGF) and acts as a co-receptor for VEGF receptor 2 to modulate EC migration, proliferation, and tube formation during postnatal and tumour angiogenesis. However, whether these SCUBE genes cooperate in modulating VEGF signalling during embryonic vascular development remains unknown. METHODS AND RESULTS: To further dissect the genetic interactions of these scube genes, transcription activator-like effector nuclease-mediated genome editing was used to generate knockout (KO) alleles of each scube gene. No overt vascular phenotypes were seen in any single scube KO mutants because of compensation by other scube genes during zebrafish development. However, scube1 and scube2 double KO (DKO) severely impaired EC filopodia extensions, migration, and proliferation, thus disrupting proper vascular lumen formation during vasculogenesis and angiogenesis as well as development of the organ-specific intestinal vasculature. Further genetic, biochemical, and molecular analyses revealed that Scube1 and Scube2 might act cooperatively at the cell-surface receptor level to facilitate Vegfa signalling during zebrafish embryonic vascularization. CONCLUSIONS: We showed for the first time that cooperation between scube1 and scube2 is critical for proper regulation of angiogenic cell behaviours and formation of functional vessels during zebrafish embryonic development.


Asunto(s)
Factor A de Crecimiento Endotelial Vascular , Pez Cebra , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Células Endoteliales/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA