Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Cell Rep ; 40(2): 111038, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35830803

RESUMEN

Despite the fundamental roles of TGF-ß family signaling in cell fate determination in all metazoans, the mechanism by which these signals are spatially and temporally interpreted remains elusive. The cell-context-dependent function of TGF-ß signaling largely relies on transcriptional regulation by SMAD proteins. Here, we discover that the DNA repair-related protein, HMCES, contributes to early development by maintaining nodal/activin- or BMP-signaling-regulated transcriptional network. HMCES binds with R-SMAD proteins, co-localizing at active histone marks. However, HMCES chromatin occupancy is independent on nodal/activin or BMP signaling. Mechanistically, HMCES competitively binds chromatin to limit binding by R-SMAD proteins, thereby forcing their dissociation and resulting in repression of their regulatory effects. In Xenopus laevis embryo, hmces KD causes dramatic development defects with abnormal left-right axis asymmetry along with increasing expression of lefty1. These findings reveal HMCES transcriptional regulatory function in the context of TGF-ß family signaling.


Asunto(s)
Activinas , Proteínas Morfogenéticas Óseas , Activinas/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Cromatina , Regulación del Desarrollo de la Expresión Génica , Células Madre Embrionarias de Ratones/metabolismo , Proteínas Smad Reguladas por Receptores/genética , Proteínas Smad Reguladas por Receptores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
2.
Biol Direct ; 15(1): 16, 2020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-33028367

RESUMEN

BACKGROUND: Amelogenesis imperfecta (AI) is a type of hereditary diseases that manifest defects in the formation or mineralization of enamel. Recently, it is reported that inactivation of FAM20C, a well-known Golgi casein kinase, caused AI. However, the mechanism of it is still unknown. The aim of this study was to explore the molecular mechanism of AI, which caused by ablation of FAM20C. RESULTS: In the Sox2-Cre;Fam20Cfl/fl (cKO) mouse, we found abnormal differentiation of ameloblasts, improper formation and mineralization of enamel, and downregulation of both mRNA and protein level of enamel matrix proteins, including amelogenin (AMEL), ameloblastin (AMBN) and enamelin (ENAM). The levels of BMP2, BMP4 and BMP7, the ligands of BMP signaling pathway, and phosphorylation of Smad1/5/8, the key regulators of BMP signaling pathway, were all decreased in the enamel matrix and the ameloblast of the cKO mice, respectively. The expression of cyclin-dependent kinase inhibitor (P21), muscle segment homeobox genes 2 (Msx2), which are the target genes of the BMP signaling pathway, and laminin 3, the downstream factor of Msx2, were all significantly decreased in the ameloblasts of the cKO mice compared to the control mice. CONCLUSION: the results of our study suggest that ablation of FAM20C leads to AI through inhibiting the Smad dependent BMP signaling pathway in the process of amelogenesis.


Asunto(s)
Amelogénesis Imperfecta/genética , Proteínas Morfogenéticas Óseas/genética , Proteínas de Unión al Calcio/genética , Proteínas del Esmalte Dental/genética , Proteínas de la Matriz Extracelular/genética , Transducción de Señal , Proteínas Smad Reguladas por Receptores/genética , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas del Esmalte Dental/metabolismo , Regulación hacia Abajo , Proteínas de la Matriz Extracelular/metabolismo , Ratones , Ratones Noqueados , Proteínas Smad Reguladas por Receptores/metabolismo
3.
Development ; 147(10)2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32345746

RESUMEN

Synapses exhibit an astonishing degree of adaptive plasticity in healthy and disease states. We have investigated whether synapses also adjust to life stages imposed by novel developmental programs for which they were never molded by evolution. Under conditions in which Drosophila larvae are terminally arrested, we have characterized synaptic growth, structure and function at the neuromuscular junction (NMJ). Although wild-type larvae transition to pupae after 5 days, arrested third instar (ATI) larvae persist for 35 days, during which time NMJs exhibit extensive overgrowth in muscle size, presynaptic release sites and postsynaptic glutamate receptors. Remarkably, despite this exuberant growth, stable neurotransmission is maintained throughout the ATI lifespan through a potent homeostatic reduction in presynaptic neurotransmitter release. Arrest of the larval stage in stathmin mutants also reveals a degree of progressive instability and neurodegeneration that was not apparent during the typical larval period. Hence, an adaptive form of presynaptic depression stabilizes neurotransmission during an extended developmental period of unconstrained synaptic growth. More generally, the ATI manipulation provides a powerful system for studying neurodegeneration and plasticity across prolonged developmental timescales.


Asunto(s)
Drosophila/crecimiento & desarrollo , Drosophila/genética , Larva/crecimiento & desarrollo , Larva/genética , Depresión Sináptica a Largo Plazo/genética , Degeneración Nerviosa/genética , Unión Neuromuscular/crecimiento & desarrollo , Animales , Axones/patología , Proteínas de Drosophila/genética , Femenino , Homeostasis/genética , Masculino , Mutación , Unión Neuromuscular/metabolismo , Interferencia de ARN , Proteínas Smad Reguladas por Receptores/genética , Estatmina/genética , Sinapsis/metabolismo , Transmisión Sináptica/genética
4.
Parasit Vectors ; 13(1): 164, 2020 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-32245505

RESUMEN

BACKGROUND: Smad proteins are essential cellular mediators within the transforming growth factor-ß (TGF-ß) superfamily. They directly transmit incoming signals from the cell surface receptors to the nucleus. In spite of their functional importance, almost nothing is known about Smad proteins in parasitic nematodes including Haemonchus contortus, an important blood-sucking nematode of small ruminants. METHODS: Based on genomic and transcriptome data for H. contortus and using bioinformatics methods, a Smad homologue (called Hco-daf-8) was inferred from H. contortus and the structural characteristics of this gene and its encoded protein Hco-DAF-8 established. Using real-time PCR and immunofluorescence assays, temporal transcriptional and spatial expression profiles of Hco-daf-8 were studied. Gene rescue in Caenorhabditis elegans was then applied to assess the function of Hco-daf-8 and a specific inhibitor of human Smad3 (called SIS3) was employed to evaluate the roles of Hco-DAF-8 in H. contortus development. RESULTS: The features of Hco-DAF-8 (502 amino acids), including conserved R-Smad domains and residues of the L3-loop that determine pathway specificity, are consistent with a TGF-ß type I receptor-activated R-Smad. The Hco-daf-8 gene was transcribed in all developmental stages of H. contortus studied, with a higher level of transcription in the fourth-stage larval (L4) females and the highest level in adult males. Hco-DAF-8 was expressed in the platymyarian muscular cells, intestine and reproductive system of adult stages. Gene rescue experiments showed that Hco-daf-8 was able to partially rescue gene function in a daf-8 deficient mutant strain of C. elegans, leading to a resumption of normal development. In H. contortus, SIS3 was shown to affect H. contortus development from the exsheathed third-stage larvae (L3s) to L4s in vitro. CONCLUSIONS: These findings suggest that Hco-DAF-8, encoded by the gene Hco-daf-8, is an important cellular mediator of H. contortus development via the TGF-ß signalling pathway. They provide a basis for future explorations of Hco-DAF-8 and associated pathways in H. contortus and other important parasitic nematodes.


Asunto(s)
Haemonchus/genética , Proteínas del Helminto/genética , Proteínas Smad Reguladas por Receptores/genética , Transcriptoma , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Biología Computacional , Femenino , Perfilación de la Expresión Génica , Genómica , Haemonchus/crecimiento & desarrollo , Masculino , Alineación de Secuencia , Transducción de Señal , Proteínas Smad Reguladas por Receptores/clasificación
5.
Apoptosis ; 24(11-12): 972-989, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31641960

RESUMEN

Developmentally regulated programmed cell death (PCD) is one of the key cellular events for precise controlling of neuronal population during postembryonic development of the central nervous system. Previously we have shown that a group of corazonin-producing peptidergic neurons (vCrz) undergo apoptosis in response to ecdysone signaling via ecdysone receptor (EcR)-B isoforms and Ultraspiracle during early phase of metamorphosis. Further utilizing genetic, transgenic, and mosaic analyses, we have found that TGF-ß signaling mediated by a glia-produced ligand, Myoglianin, type-I receptor Baboon (particularly Babo-A isoform) and dSmad2, is also required autonomously for PCD of the vCrz neurons. Our studies show that TGF-ß signaling is not acting epistatically to EcR or vice versa. We also show that ectopic expression of a constitutively active phosphomimetic form of dSmad2 (dSmad2PM) is capable of inducing premature death of vCrz neurons in larva but not other larval neurons. Intriguingly, the dSmad2PM-mediated killing is completely suppressed by coexpression of a dominant-negative form of EcR (EcRDN), suggesting that EcR function is required for the proapoptotic dSmad2PM function. Based on these data, we suggest that TGF-ß and ecdysone signaling pathways act cooperatively to induce vCrz neuronal PCD. We propose that this type of two-factor authentication is a key developmental strategy to ensure the timely PCD of specific larval neurons during metamorphosis.


Asunto(s)
Receptores de Activinas/metabolismo , Apoptosis , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Metamorfosis Biológica/genética , Neuronas/metabolismo , Receptores de Esteroides/metabolismo , Receptores de Activinas/genética , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Animales , Animales Modificados Genéticamente , Apoptosis/fisiología , Sistema Nervioso Central/citología , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Ecdisona/metabolismo , Ecdisona/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Larva/citología , Larva/metabolismo , Metamorfosis Biológica/fisiología , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuropéptidos/genética , Neuropéptidos/metabolismo , Isoformas de Proteínas/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores de Esteroides/genética , Transducción de Señal/genética , Proteínas Smad Reguladas por Receptores/genética , Proteínas Smad Reguladas por Receptores/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/fisiología
6.
Cell Cycle ; 18(15): 1714-1726, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31234714

RESUMEN

Objective: The present study was conducted to determine the role of gremlin during the development of posterior capsular opacification (PCO) via in vitro and in vivo experiments. Methods: The activation, roles and relationships of the BMPs/Smad1/5, MAPK, FAK and AKT signaling pathways in human lens epithelial cells (HLECs) after gremlin induction were detected by western blotting and real-time PCR. Wound-healing, transwell, capsular bag models and rat PCO models assays were used to test the effects of gremlin on HLECs' migration, proliferation, EMT-specific protein α-smooth muscle actin(α-SMA)and development of PCO in rats. Results: Our data showed that knockdown of the gremlin inhibited the development of PCO and reduced expression of α-SMA in rats. While gremlin did not alter the migration of HLECs, it increased the expression of p-ERK and p-AKT. Knockout of Smad2 or Smad3 inhibited the expression of p-ERK and p-AKT proteins induced by gremlin. Gremlin also reduced BMP4-induced expression of the p-Smad1/5 protein. Finally, knockout of Smad1/5 increased gremlin-induced expression of α-SMA, fibronectin and type I collagen (COL-1) in HLECs. Conclusion: These results suggested that gremlin contributed to the development of PCO by promoting LEC proliferation, activation of TGF-ß/Smad, ERK and AKT signaling and inhibition of BMPs/Smad1/5 signaling. Furthermore, inhibiting gremlin effectively impaired both PCO development in rats and EMT in the lens capsule. Thus, our data suggest that gremlin might be a potential target for PCO.


Asunto(s)
Opacificación Capsular/metabolismo , Células Epiteliales/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas/metabolismo , Actinas/metabolismo , Animales , Opacificación Capsular/genética , Línea Celular , Movimiento Celular/genética , Proliferación Celular/genética , Colágeno Tipo I/metabolismo , Transición Epitelial-Mesenquimal/genética , Femenino , Fibronectinas/metabolismo , Silenciador del Gen , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/farmacología , Sistema de Señalización de MAP Quinasas/genética , Masculino , Proteínas/genética , ARN Interferente Pequeño , Ratas , Proteínas Smad Reguladas por Receptores/genética , Proteínas Smad Reguladas por Receptores/metabolismo , Porcinos , Factor de Crecimiento Transformador beta2/metabolismo , Cicatrización de Heridas/genética
7.
Evol Dev ; 21(1): 44-55, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30588766

RESUMEN

Members of the phylum Arthropoda, comprising over 80% of total animal species, have evolved regenerative abilities, but little is known about the molecular mechanisms mediating this process. Transforming growth factor ß (TGF-ß) signaling mediates a diverse set of essential processes in animals and is a good candidate pathway for regulation of regeneration in arthropods. In this study we investigated the role of activin signaling, a TGF-ß superfamily pathway, in limb regeneration in the crayfish. We identified and cloned a downstream transcription factor in the activin pathway, Smox, and characterized its function with regard to other elements of the activin signaling pathway. Gene knockdown of Smox by RNAi induced regeneration of complete but smaller pereopods after autotomy. This indicates that activin signaling via Smox functions in regulation of pereopod growth and size. The expression levels of both Smox and the activin receptor babo were closely correlated with molting. The expression level of Smox increased when babo was knocked down by RNAi, indicating that Smox and babo transcription are linked. Our study suggests that the Babo-Smox system in activin signaling is conserved in decapods, and supports an evolutionary conservation of this aspect of molecular signaling during regeneration between protostomes and deuterostomes.


Asunto(s)
Astacoidea/fisiología , Proteínas Smad Reguladas por Receptores/metabolismo , Animales , Clonación Molecular , Extremidades/fisiología , Técnicas de Silenciamiento del Gen , Regeneración , Proteínas Smad Reguladas por Receptores/química , Proteínas Smad Reguladas por Receptores/genética
8.
Arch Toxicol ; 92(9): 2935-2946, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30097701

RESUMEN

MicroRNA (miRNA)-mediated gene regulation contributes to liver pathophysiology, including hepatic stellate cell (HSC) activation and fibrosis progression. Here, we investigated the role of miR-942 in human liver fibrosis. The expression of miR-942, HSC activation markers, transforming growth factor-beta pseudoreceptor BMP and activin membrane-bound inhibitor (BAMBI), as well as collagen deposition, were investigated in 100 liver specimens from patients with varying degree of hepatitis B virus (HBV)-related fibrosis. Human primary HSCs and the immortalized cell line (LX2 cells) were used for functional studies. We found that miR-942 expression was upregulated in activated HSCs and correlated inversely with BAMBI expression in liver fibrosis progression. Transforming growth factor beta (TGF-ß) and lipopolyssacharide (LPS), two major drivers of liver fibrosis and inflammation, induce miR-942 expression in HSCs via Smad2/3 respective NF-κB/p50 binding to the miR-942 promoter. Mechanistically, the induced miR-942 degrades BAMBI mRNA in HSCs, thereby sensitizing the cells for fibrogenic TGF-ß signaling and also partly mediates LPS-induced proinflammatory HSC fate. In conclusion, the TGF-ß and LPS-induced miR-942 mediates HSC activation through downregulation of BAMBI in human liver fibrosis. Our study provides new insights on the molecular mechanism of HSC activation and fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/patología , Cirrosis Hepática/patología , Proteínas de la Membrana/genética , MicroARNs/metabolismo , Células Cultivadas , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Humanos , Lipopolisacáridos/farmacología , Cirrosis Hepática/genética , Proteínas de la Membrana/metabolismo , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Proteínas Smad Reguladas por Receptores/genética , Proteínas Smad Reguladas por Receptores/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
9.
Biomed Pharmacother ; 97: 429-438, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29091893

RESUMEN

AIM: Aquaporins (AQPs) are water-channels that play important roles in brain water homeostasis and cerebral edema induced by brain injury. This study aimed to investigate the relationship between AQP4, bone morphogenetic protein 4 (BMP4)/Smad1/5/8 signaling pathway and isoflurane post-conditiong, which has effects on brain edema in rats with cerebral ischemia/reperfusion (I/R) injury. METHODS: Cerebral I/R injury was induced in rats by using the middle cerebral artery occlusion (MCAO) model for 90min, followed by 24h of reperfusion. Isoflurane post-conditioning (ISO) group received 90min ischemia and underwent 1.5% isoflurane post-conditioning for 60min after initiating reperfusion. Neurobehavior, brain water content, thionine staining and 2, 3, 5-triphenyl tetrazolium chloride staining were evaluated to measure levels of brain edema and damage. Expressions of AQP4, BMP4, Smad1/5/8 and phosphorylated Smad1/5/8 were detected by using Western blot, quantitative real-time polymerase chain reaction (qRT-PCR), and immunofluorescence (IF) staining. RESULTS: Compared with the Sham group, neurological behavior score, brain infarct volume and water content of MCAO model rats increased with reperfusion injury. However, in the ISO group, cell edema and damage of brain was significantly ameliorated (P<0.01). qRT-PCR showed less AQP4 mRNA expression in the hippocampal tissue of the ISO group than in the I/R group (P<0.01). Western blot and immunofluorescence results showed similar changes in protein levels of both groups. Related protein expressions showed expressions of BMP4 and Smad1/5/8 increased in the ISO group (P<0.01), whereas total Smad1/5/8 expression didn't change in all groups. When BMP4 inhibitor (LDN193189) was injected, expression levels of AQP4 increased and neuronal density decreased (P<0.05). By contrast, expression levels of BMP4 did not change significantly after pre-injection of AQP4 inhibitor (TGN020) (P>0.05), but neuronal density increased (P<0.05). CONCLUSION: Isoflurane post-conditioning may inhibit occurrence of brain edema and reduce cerebral I/R injury through down-regulating expression of AQP4, This process may be related to the activation of BMP4/Smad1/5/8 signaling pathway.


Asunto(s)
Acuaporina 4/biosíntesis , Proteína Morfogenética Ósea 4/biosíntesis , Isquemia Encefálica/metabolismo , Poscondicionamiento Isquémico/métodos , Isoflurano/administración & dosificación , Daño por Reperfusión/metabolismo , Proteínas Smad Reguladas por Receptores/biosíntesis , Animales , Acuaporina 4/antagonistas & inhibidores , Acuaporina 4/genética , Proteína Morfogenética Ósea 4/genética , Isquemia Encefálica/terapia , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Expresión Génica , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/terapia , Masculino , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/terapia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteínas Smad Reguladas por Receptores/genética , Proteína Smad1/biosíntesis , Proteína Smad1/genética , Proteína Smad5/biosíntesis , Proteína Smad5/genética , Proteína Smad8/biosíntesis , Proteína Smad8/genética
10.
Reproduction ; 153(2): 221-231, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27879343

RESUMEN

The molecular mechanisms involved in regulating the development of small, gonadotrophin-independent follicles are poorly understood; however, many studies have highlighted an essential role for TGFB ligands. Canonical TGFB signalling is dependent upon intracellular SMAD proteins that regulate transcription. STRAP has been identified in other tissues as an inhibitor of the TGFB-SMAD signalling pathway. Therefore, in this study we aimed to determine the expression and role of STRAP in the context of early follicle development. Using qPCR, Strap, Smad3 and Smad7 revealed similar expression profiles in immature ovaries from mice aged 4-16 days containing different populations of early growing follicles. STRAP and SMAD2/3 proteins co-localised in granulosa cells of small follicles using immunofluorescence. Using an established culture model, neonatal mouse ovary fragments with a high density of small non-growing follicles were used to examine the effects of Strap knockdown using siRNA and STRAP protein inhibition by immuno-neutralisation. Both interventions caused a reduction in the proportion of small, non-growing follicles and an increase in the proportion and size of growing follicles in comparison to untreated controls, suggesting inhibition of STRAP facilitates follicle activation. Recombinant STRAP protein had no effect on small, non-growing follicles, but increased the mean oocyte size of growing follicles in the neonatal ovary model and also promoted the growth of isolated preantral follicles in vitro Overall findings indicate STRAP is expressed in the mouse ovary and is capable of regulating development of small follicles in a stage-dependent manner.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Folículo Ovárico/crecimiento & desarrollo , Ovario/crecimiento & desarrollo , Proteínas Adaptadoras Transductoras de Señales/análisis , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Femenino , Técnicas de Silenciamiento del Gen , Células de la Granulosa/química , Ratones , Ratones Endogámicos C57BL , Oocitos/efectos de los fármacos , Oocitos/crecimiento & desarrollo , Ovario/metabolismo , Proteínas de Unión al ARN , Proteínas Recombinantes/farmacología , Transducción de Señal/fisiología , Proteínas Smad Reguladas por Receptores/genética , Proteína Smad2/análisis , Proteína smad3/análisis , Proteína smad3/genética , Transcriptoma , Factor de Crecimiento Transformador beta/metabolismo
11.
Mol Nutr Food Res ; 60(12): 2587-2601, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27506630

RESUMEN

SCOPE: Yuja (Citrus junos Tanaka) possesses various health benefits, but its effects on bone health are unknown. In this study, the preventative effects of yuja peel ethanol extract (YPEE) on osteopenia were determined in ovariectomized (OVX) rats, and the mechanisms by which YPEE and its flavanones regulate osteoblastogenesis were examined in vitro. METHODS AND RESULTS: The effects of YPEE on osteoblastogenesis were investigated in MC3T3-E1 cells. YPEE promoted alkaline phosphatase (ALP) activity, mineralization, and the expression of osteoblast differentiation marker genes, such as ALP, runt-related transcription factor 2 (Runx2), and osteocalcin. YPEE and its flavanones promoted osteoblast differentiation via BMP-2-mediated p38 and the Smad1/5/8 signaling pathway. YPEE supplementation significantly decreased body weight and increased uterine weight and bone mineral density in OVX rats. Based on a micro-CT analysis of femurs, YPEE significantly attenuated osteopenia and increased trabecular volume fraction, trabecular separation, and trabecular number (p < 0.05). CONCLUSION: Dietary YPEE has a protective effect on OVX-induced osteopenia. YPEE and its flavanones promote osteoblastogenesis via the activation of the BMP/p38/Smad/Runx2 pathways. These results extend our knowledge of the beneficial effects of YPEE and provide a basis for the development of novel therapies for osteoporosis.


Asunto(s)
Enfermedades Óseas Metabólicas/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Flavanonas/farmacología , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Extractos Vegetales/farmacología , Células 3T3 , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Densidad Ósea/efectos de los fármacos , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Citrus/química , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Dislipidemias/tratamiento farmacológico , Dislipidemias/etiología , Femenino , Ratones , Osteoblastos/citología , Osteocalcina/genética , Osteocalcina/metabolismo , Ovariectomía , Células RAW 264.7 , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteínas Smad Reguladas por Receptores/genética , Proteínas Smad Reguladas por Receptores/metabolismo
12.
Dev Biol ; 417(1): 104-13, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27452629

RESUMEN

In contrast with Drosophila melanogaster, practically nothing is known about the involvement of the TGF-ß signaling pathway in the metamorphosis of hemimetabolan insects. To partially fill this gap, we have studied the role of Smad factors in the metamorphosis of the German cockroach, Blattella germanica. In D. melanogaster, Mad is the canonical R-Smad of the BMP branch of the TGF-ß signaling pathway, Smox is the canonical R-Smad of the TGF-ß/Activin branch and Medea participates in both branches. In insects, metamorphosis is regulated by the MEKRE93 pathway, which starts with juvenile hormone (JH), whose signal is transduced by Methoprene-tolerant (Met), which stimulates the expression of Krüppel homolog 1 (Kr-h1) that acts to repress E93, the metamorphosis trigger. In B. germanica, metamorphosis is determined at the beginning of the sixth (final) nymphal instar (N6), when JH production ceases, the expression of Kr-h1 declines, and the transcription of E93 begins to increase. The RNAi of Mad, Smox and Medea in N6 of B. germanica reveals that the BMP branch of the TGF-ß signaling pathway regulates adult ecdysis and wing extension, mainly through regulating the expression of bursicon, whereas the TGF-ß/Activin branch contributes to increasing E93 and decreasing Kr-h1 at the beginning of N6, crucial for triggering adult morphogenesis, as well as to regulating the imaginal molt timing.


Asunto(s)
Cucarachas/embriología , Drosophila melanogaster/embriología , Metamorfosis Biológica/fisiología , Muda/fisiología , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Activinas/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hormonas Juveniles/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Transducción de Señal , Proteínas Smad Reguladas por Receptores/genética , Proteínas Smad Reguladas por Receptores/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
Sci Rep ; 6: 25594, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27146698

RESUMEN

Cell cycle regulation is critical for chondrocyte differentiation and hypertrophy. Recently we identified the Notch signaling pathway as an important regulator of chondrocyte proliferation and differentiation during mouse cartilage development. To investigate the underlying mechanisms, we assessed the role for Notch signaling regulation of the cell cycle during chondrocyte differentiation. Real-time RT-PCR data showed that over-expression of the Notch Intracellular Domain (NICD) significantly induced the expression of p57, a cell cycle inhibitor, in chondrocytes. Flow cytometric analyses further confirmed that over-expression of NICD in chondrocytes enhances the G0/G1 cell cycle transition and cell cycle arrest. In contrast, treatment of chondrocytes with the Notch inhibitor, DAPT, decreased both endogenous and BMP2-induced SMAD 1/5/8 phosphorylation and knockdown of SMAD 1/5/8 impaired NICD-induced chondrocyte differentiation and p57 expression. Co-immunoprecipitation using p-SMAD 1/5/8 and NICD antibodies further showed a strong interaction of these proteins during chondrocyte maturation. Finally, RT-PCR and Western blot results revealed a significant reduction in the expression of the SMAD-related phosphatase, PPM1A, following NICD over-expression. Taken together, our results demonstrate that Notch signaling induces cell cycle arrest and thereby initiates chondrocyte hypertrophy via BMP/SMAD-mediated up-regulation of p57.


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Puntos de Control del Ciclo Celular/genética , Condrocitos/metabolismo , Receptores Notch/genética , Animales , Proteína Morfogenética Ósea 2/metabolismo , Diferenciación Celular/genética , Aumento de la Célula , Línea Celular Tumoral , Proliferación Celular/genética , Condrocitos/patología , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación de la Expresión Génica , Ratones , Fosforilación , Interferencia de ARN , Receptores Notch/metabolismo , Transducción de Señal/genética , Proteínas Smad Reguladas por Receptores/genética , Proteínas Smad Reguladas por Receptores/metabolismo
14.
Arterioscler Thromb Vasc Biol ; 36(3): 553-60, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26769046

RESUMEN

OBJECTIVE: Bone morphogenic protein 4 (BMP4) is an important mediator of endothelial dysfunction in cardio-metabolic diseases, whereas platelet-derived growth factors (PDGFs) are major angiogenic and proinflammatory mediator, although the functional link between these 2 factors is unknown. The present study investigated whether PDGF mediates BMP4-induced endothelial dysfunction in diabetes mellitus. APPROACH AND RESULTS: We generated Ad-Bmp4 to overexpress Bmp4 and Ad-Pdgfa-shRNA to knockdown Pdgfa in mice through tail intravenous injection. SMAD4-shRNA lentivirus, SMAD1-shRNA, and SMAD5 shRNA adenovirus were used for knockdown in human and mouse endothelial cells. We found that PDGF-AA impaired endothelium-dependent vasodilation in aortas and mesenteric resistance arteries. BMP4 upregulated PDGF-AA in human and mouse endothelial cells, which was abolished by BMP4 antagonist noggin or knockdown of SMAD1/5 or SMAD4. BMP4-impared relaxation in mouse aorta was also ameliorated by PDGF-AA neutralizing antibody. Tail injection of Ad-Pdgfa-shRNA ameliorates endothelial dysfunction induced by Bmp4 overexpression (Ad-Bmp4) in vivo. Serum PDGF-AA was elevated in both diabetic patients and diabetic db/db mice compared with nondiabetic controls. Pdgfa-shRNA or Bmp4-shRNA adenovirus reduced serum PDGF-AA concentration in db/db mice. PDGF-AA neutralizing antibody or tail injection with Pdgfa-shRNA adenovirus improved endothelial function in aortas and mesenteric resistance arteries from db/db mice. The effect of PDGF-AA on endothelial function in mouse aorta was also inhibited by Ad-Pdgfra-shRNA to inhibit PDGFRα. CONCLUSIONS: The present study provides novel evidences to show that PDGF-AA impairs endothelium-dependent vasodilation and PDGF-AA mediates BMP4-induced adverse effect on endothelial cell function through SMAD1/5- and SMAD4-dependent mechanisms. Inhibition of PGDF-AA ameliorates vascular dysfunction in diabetic mice.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Diabetes Mellitus/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Smad Reguladas por Receptores/metabolismo , Vasodilatación , Adulto , Anciano , Animales , Anticuerpos Neutralizantes/farmacología , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/farmacología , Estudios de Casos y Controles , Células Cultivadas , Diabetes Mellitus/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/farmacología , Interferencia de ARN , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Smad Reguladas por Receptores/genética , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Transfección , Regulación hacia Arriba , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
15.
Stem Cell Reports ; 6(1): 85-94, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26711875

RESUMEN

Naive mouse embryonic stem cells (mESCs) are in a metastable state and fluctuate between inner cell mass- and epiblast-like phenotypes. Here, we show transient activation of the BMP-SMAD signaling pathway in mESCs containing a BMP-SMAD responsive reporter transgene. Activation of the BMP-SMAD reporter transgene in naive mESCs correlated with lower levels of genomic DNA methylation, high expression of 5-methylcytosine hydroxylases Tet1/2 and low levels of DNA methyltransferases Dnmt3a/b. Moreover, naive mESCs, in which the BMP-SMAD reporter transgene was activated, showed higher resistance to differentiation. Using double Smad1;Smad5 knockout mESCs, we showed that BMP-SMAD signaling is dispensable for self-renewal in both naive and ground state. These mutant mESCs were still pluripotent, but they exhibited higher levels of DNA methylation than their wild-type counterparts and had a higher propensity to differentiate. We showed that BMP-SMAD signaling modulates lineage priming in mESCs, by transiently regulating the enzymatic machinery responsible for DNA methylation.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Linaje de la Célula/fisiología , Autorrenovación de las Células/fisiología , Células Madre Embrionarias de Ratones/metabolismo , Transducción de Señal/fisiología , Proteínas Smad Reguladas por Receptores/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Linaje de la Célula/genética , Autorrenovación de las Células/genética , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Perfilación de la Expresión Génica/métodos , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Células Madre Embrionarias de Ratones/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Proteínas Smad Reguladas por Receptores/genética , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo
16.
ACS Chem Neurosci ; 7(1): 90-9, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26505647

RESUMEN

Identification of small molecules that direct neural stem cells (NSCs) into specific cell types would be helpful to understand the molecular mechanisms involved in regulation of NSC fate, and facilitate the development of therapeutic applications. In the current study, we developed and screened small molecules that can modulate the fate of NSCs that are derived from rat fetal cortex. Among these compounds, compounds 5 and 6 successfully differentiated NSCs into astrocytes and neurons, respectively. Compound 5 induced astrocytogenesis by increasing expression of interleukin-6, bone morphogenetic protein 2 and leukemia inhibitory factor and through consequent phosphorylation of signal transducer and activator of transcription 3 and Sma- and Mad-related protein 1/5/8 in NSCs. In addition, compound 5 increased the expression of fibroblast growth factor (FGF) 2 and FGF8 which may regulate the branching and morphology of astrocytes. Taken together, our results suggest that these small molecules can serve as a useful tool to study cell fate determination in NSCs and be used as an inexpensive alternative to cytokines to study mechanisms of astrocytogenesis.


Asunto(s)
Astrocitos/efectos de los fármacos , Citocinas/farmacología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Células-Madre Neurales/efectos de los fármacos , Organogénesis/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Smad Reguladas por Receptores/metabolismo , Animales , Recuento de Células , Células Cultivadas , Embrión de Mamíferos , Proteína Quinasa 3 Activada por Mitógenos/genética , Modelos Moleculares , Células-Madre Neurales/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/genética , Proteínas Smad Reguladas por Receptores/genética , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo , Proteína Smad8/genética , Proteína Smad8/metabolismo
17.
Arterioscler Thromb Vasc Biol ; 35(12): 2626-37, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26471266

RESUMEN

OBJECTIVE: Bone morphogenetic protein-9 (BMP9)/activin-like kinase-1 and delta-like 4 (DLL4)/Notch promote endothelial quiescence, and we aim to understand mechanistic interactions between the 2 pathways. We identify new targets that contribute to endothelial quiescence and test whether loss of Dll4(+/-) in adult vasculature alters BMP signaling. APPROACH AND RESULTS: Human endothelial cells respond synergistically to BMP9 and DLL4 stimulation, showing complete quiescence and induction of HEY1 and HEY2. Canonical BMP9 signaling via activin-like kinase-1-Smad1/5/9 was disrupted by inhibition of Notch signaling, even in the absence of exogenous DLL4. Similarly, DLL4 activity was suppressed when the basal activin-like kinase-1-Smad1/5/9 pathway was inhibited, showing that these pathways are interdependent. BMP9/DLL4 required induction of P27(KIP1) for quiescence, although multiple factors are involved. To understand these mechanisms, we used proteomics data to identify upregulation of thrombospondin-1, which contributes to the quiescence phenotype. To test whether Dll4 regulates BMP/Smad pathways and endothelial cell phenotype in vivo, we characterized the vasculature of Dll4(+/-) mice, analyzing endothelial cells in the lung, heart, and aorta. Together with changes in endothelial structure and vascular morphogenesis, we found that loss of Dll4 was associated with a significant upregulation of pSmad1/5/9 signaling in lung endothelial cells. Because steady-state endothelial cell proliferation rates were not different in the Dll4(+/-) mice, we propose that the upregulation of pSmad1/5/9 signaling compensates to maintain endothelial cell quiescence in these mice. CONCLUSIONS: DLL4/Notch and BMP9/activin-like kinase-1 signaling rely on each other's pathways for full activity. This represents an important mechanism of cross talk that enhances endothelial quiescence and sensitively coordinates cellular responsiveness to soluble and cell-tethered ligands.


Asunto(s)
Senescencia Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Endoteliales/metabolismo , Factores de Diferenciación de Crecimiento/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Receptor Notch1/metabolismo , Trombospondina 1/metabolismo , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Aorta/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Unión al Calcio , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Células Cultivadas , Vasos Coronarios/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Genotipo , Factor 2 de Diferenciación de Crecimiento , Humanos , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Pulmón/irrigación sanguínea , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Interferencia de ARN , Receptor Notch1/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Proteínas Smad Reguladas por Receptores/genética , Proteínas Smad Reguladas por Receptores/metabolismo , Trombospondina 1/genética , Transfección
18.
Am J Rhinol Allergy ; 29(5): e152-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26265084

RESUMEN

BACKGROUND: Chronic rhinosinusitis (CRS) without nasal polyps (CRSsNP) and with nasal polyps (CRSwNP) is reported to involve different inflammatory processes in sinonasal mucosa and bone tissue, and these processes remain uncharacterized. OBJECTIVE: We aimed to investigate the molecular mechanisms of osteitis in Chinese patients with CRS to better understand the pathogenesis of CRS. METHODS: The study included 10 controls, 16 patients with CRSsNP, and 23 patients with CRSwNP. Ethmoid bone tissue samples were evaluated by histologic examination. Quantitative real-time reverse transcription polymerase chain reaction was used to assess expression of transforming growth factor (TGF) ß1, TGF-ß receptor I and II, Smad2, and Smad3. Immunohistochemical examination of osteoblast expression of TGF-ß1, TGF-ß receptor I and II, phosphorylated (p) Smad2, and p-Smad3 in ethmoid bone tissue was also performed. RESULTS: The histopathologic evaluation of ethmoid sinus bone tissue showed that eosinophils had infiltrated the periosteum and induced TGF-ß1 expression, periosteal thickening, increased osteoblast activity, and neo-osteogenesis. Messenger RNA levels of TGF-ß1, TGF-ß receptor I, and Smad3 in CRSwNP ethmoid bone tissues were significantly higher than those in ethmoid bone tissues of patients with CRSsNP and the controls. Immunohistochemical staining showed that TGF-ß1, TGF-ß receptor I, p-Smad2, and p-Smad3 protein expression was upregulated in patients with CRSwNP, consistent with the corresponding messenger RNA levels. CONCLUSION: Different signaling pathways are involved in osteitis in CRS and are activated by the TGF-ß/Smad signaling pathway in CRSwNP versus the TGF-ß/Smad-independent signaling pathway in CRSsNP. Eosinophil infiltration of the periosteum, along with TGF-ß1 expression, in CRSwNP indicates that eosinophils may play an important role in the bone remodeling process in CRSwNP.


Asunto(s)
Regulación de la Expresión Génica , Pólipos Nasales/genética , Osteítis/genética , Rinitis/genética , Sinusitis/genética , Proteínas Smad Reguladas por Receptores/genética , Factor de Crecimiento Transformador beta/genética , Adulto , Enfermedades Óseas Infecciosas/complicaciones , Enfermedades Óseas Infecciosas/genética , Enfermedades Óseas Infecciosas/metabolismo , Enfermedad Crónica , Hueso Etmoides , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Pólipos Nasales/complicaciones , Pólipos Nasales/metabolismo , Osteítis/complicaciones , Osteítis/metabolismo , ARN/genética , Estudios Retrospectivos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rinitis/complicaciones , Rinitis/metabolismo , Transducción de Señal , Sinusitis/complicaciones , Sinusitis/metabolismo , Proteínas Smad Reguladas por Receptores/biosíntesis , Proteína Smad2/biosíntesis , Proteína Smad2/genética , Proteína smad3/biosíntesis , Proteína smad3/genética , Factor de Crecimiento Transformador beta/biosíntesis , Adulto Joven
19.
Stem Cells Dev ; 24(11): 1320-31, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25603382

RESUMEN

A low-efficiency yield hinders the use of stem cells as a source of endothelial cells (ECs) for therapeutic vascularization, and the diversity of the transforming growth factor-ß (TGF-ß) superfamily has undermined understanding the effects of its potent vascularization-inducing. Herein, we studied the role of the TGF-ß superfamily in EC differentiation of rat bone marrow mesenchymal stem cells (MSCs) induced by Smad2/3 and Smad1/5/8 signaling. MSCs that had been sorted by flow cytometry as CD31-negative were cultured for 14 days in medium supplemented with vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) as the control. The Smad2/3 pathway was activated by TGF-ß1 and Smad1/5/8 by bone morphogenetic proteins (BMPs). In the early phase in the Smad2/3-activated group, there were 10% CD31-positive cells, which was significantly higher than in the control group. A low Smad1/5/8 phosphorylation level after BMP4 activation doubled the number of CD31-positive cells, while a higher phosphorylation level after BMP9 activation showed no effect. A Smad2/3 inhibitor initially blocked differentiation but later promoted it, while a Smad1/5/8 inhibitor reversed the induction observed with BMPs. Moreover, the positive effects of R-Smad on differentiation were weakened by the VEGF neutralizing antibody, and a Smad3 inhibitor decreased VEGF expression and blocked differentiation in both the early and late phases. In conclusion, differentiation of ECs from MSCs via Smad2/3 signaling is stage dependent. Activation, particularly by Smad3, significantly promotes differentiation at an early phase but later is suppressive. A low Smad1/5/8 phosphorylation level has a positive effect, and R-Smad effects are partly mediated by VEGF.


Asunto(s)
Diferenciación Celular , Células Endoteliales/citología , Células Madre Mesenquimatosas/metabolismo , Transducción de Señal , Proteínas Smad Reguladas por Receptores/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Células Endoteliales/metabolismo , Factor 9 de Diferenciación de Crecimiento/genética , Factor 9 de Diferenciación de Crecimiento/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Ratas , Ratas Sprague-Dawley , Proteínas Smad Reguladas por Receptores/genética , Factor A de Crecimiento Endotelial Vascular/genética
20.
PLoS One ; 9(9): e107443, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25255438

RESUMEN

Members of the TGF-ß superfamily play numerous roles in nervous system development and function. In Drosophila, retrograde BMP signaling at the neuromuscular junction (NMJ) is required presynaptically for proper synapse growth and neurotransmitter release. In this study, we analyzed whether the Activin branch of the TGF-ß superfamily also contributes to NMJ development and function. We find that elimination of the Activin/TGF-ß type I receptor babo, or its downstream signal transducer smox, does not affect presynaptic NMJ growth or evoked excitatory junctional potentials (EJPs), but instead results in a number of postsynaptic defects including depolarized membrane potential, small size and frequency of miniature excitatory junction potentials (mEJPs), and decreased synaptic densities of the glutamate receptors GluRIIA and B. The majority of the defective smox synaptic phenotypes were rescued by muscle-specific expression of a smox transgene. Furthermore, a mutation in actß, an Activin-like ligand that is strongly expressed in motor neurons, phenocopies babo and smox loss-of-function alleles. Our results demonstrate that anterograde Activin/TGF-ß signaling at the Drosophila NMJ is crucial for achieving normal abundance and localization of several important postsynaptic signaling molecules and for regulating postsynaptic membrane physiology. Together with the well-established presynaptic role of the retrograde BMP signaling, our findings indicate that the two branches of the TGF-ß superfamily are differentially deployed on each side of the Drosophila NMJ synapse to regulate distinct aspects of its development and function.


Asunto(s)
Activinas/metabolismo , Drosophila melanogaster/citología , Unión Neuromuscular/citología , Unión Neuromuscular/metabolismo , Receptores de Glutamato/metabolismo , Transducción de Señal , Potenciales Sinápticos , Activinas/genética , Animales , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Ligandos , Mutación , Transporte de Proteínas , Proteínas Smad Reguladas por Receptores/deficiencia , Proteínas Smad Reguladas por Receptores/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA