Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
2.
Nat Commun ; 13(1): 497, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35079017

RESUMEN

Morphogens are signaling molecules that convey positional information and dictate cell fates during development. Although ectopic expression in model organisms suggests that morphogen gradients form through diffusion, little is known about how morphogen gradients are created and interpreted during mammalian embryogenesis due to the combined difficulties of measuring endogenous morphogen levels and observing development in utero. Here we take advantage of a human gastruloid model to visualize endogenous Nodal protein in living cells, during specification of germ layers. We show that Nodal is extremely short range so that Nodal protein is limited to the immediate neighborhood of source cells. Nodal activity spreads through a relay mechanism in which Nodal production induces neighboring cells to transcribe Nodal. We further show that the Nodal inhibitor Lefty, while biochemically capable of long-range diffusion, also acts locally to control the timing of Nodal spread and therefore of mesoderm differentiation during patterning. Our study establishes a paradigm for tissue patterning by an activator-inhibitor pair.


Asunto(s)
Blastocisto/metabolismo , Gástrula/metabolismo , Gastrulación/genética , Células Madre Embrionarias Humanas/metabolismo , Proteína Nodal/genética , Blastocisto/citología , Línea Celular , Difusión , Técnica del Anticuerpo Fluorescente/métodos , Gástrula/citología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Células Madre Embrionarias Humanas/citología , Humanos , Hibridación Fluorescente in Situ/métodos , Factores de Determinación Derecha-Izquierda/genética , Factores de Determinación Derecha-Izquierda/metabolismo , Microscopía Confocal/métodos , Microscopía Fluorescente/métodos , Proteína Nodal/metabolismo
3.
Dev Biol ; 481: 172-178, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34737126

RESUMEN

Placentas control the maternal-fetal transport of nutrients and gases. Placental reactions to adverse intrauterine conditions affect fetal development. Such adverse conditions occur in pregnancies complicated by diabetes, leading to alterations in placental anatomy and physiology. In this study, streptozocin (STZ) injection produced sustained hyperglycemia during pregnancy in rats. Hyperglycemic pregnant rats had gained significantly less weight than normal pregnant rats on embryonic day 15.5. We investigated the influence of diabetes on placental anatomy and physiology. Compared with controls, the diabetic group had a markedly thicker junctional zone at embryonic day 15.5. To explore a mechanism for this abnormality, we examined Nodal expression in the junctional zone of control and diabetic groups. We found lower expression of Nodal in the diabetic group. We then investigated the expression of its target gene p27Kip1 (p27), which is related to cell proliferation. In vitro, Nodal overexpression up-regulated p27 protein levels while interfered EBAF up-regulated p27. In vivo, the expression of p27 was lower in diabetic compared with normal rats, and localization was similar between the two groups. In contrast, a higher expression of PCNA was found in diabetic versus normal placenta. Endometrial bleeding associated factor (EBAF), an up-stream molecular regulator of Nodal, was expressed at higher levels in placenta from diabetic versus normal rats. Based on these results, we speculate that the EBAF/Nodal/p27 signaling pathway plays a role in morphological change of diabetic placenta.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Diabetes Mellitus Experimental/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Determinación Derecha-Izquierda/metabolismo , Proteína Nodal/metabolismo , Placenta/metabolismo , Embarazo en Diabéticas/metabolismo , Transducción de Señal , Animales , Femenino , Embarazo , Ratas , Ratas Sprague-Dawley
4.
Sci Rep ; 11(1): 24016, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34907278

RESUMEN

Embryo implantation is a key step in establishing pregnancy and a major limiting factor in IVF. Implantation requires a receptive endometrium but the mechanisms governing receptivity are not well understood. We have recently discovered that podocalyxin (PCX or PODXL) is a key negative regulator of human endometrial receptivity. PCX is expressed in all endometrial epithelial cells in the non-receptive endometrium but selectively down-regulated in the luminal epithelium at receptivity. We have further demonstrated that this down-regulation is essential for implantation because PCX inhibits embryo attachment and penetration. However, how PCX confers this role is unknown. In this study, through RNAseq analysis of Ishikawa cell line stably overexpressing PCX, we discovered that PCX suppresses expression of genes controlling cell adhesion and communication, but increases those governing epithelial barrier functions, especially the adherens and tight junctions. Moreover, PCX suppresses multiple factors such as LIF and signaling pathways including Wnt and calcium signaling that support receptivity but stimulates anti-implantation genes such as LEFTY2. Functional studies confirmed that PCX promotes epithelial barrier functions by increasing key epithelial junction proteins such as E-cadherin and claudin 4. PCX thus promotes an anti-adhesive and impermeable epithelium while impedes pro-implantation factors to negatively control endometrial receptivity for implantation.


Asunto(s)
Endometrio/metabolismo , Células Epiteliales/metabolismo , Sialoglicoproteínas/metabolismo , Antígenos CD/metabolismo , Cadherinas/metabolismo , Implantación del Embrión , Femenino , Humanos , Inflamación/metabolismo , Factores de Determinación Derecha-Izquierda/metabolismo , Embarazo
5.
Invest Ophthalmol Vis Sci ; 62(15): 27, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34964803

RESUMEN

Purpose: Exfoliation syndrome (XFS) is a condition characterized by the production of insoluble fibrillar aggregates (exfoliation material; XFM) in the eye and elsewhere. Many patients with XFS progress to exfoliation glaucoma (XFG), a significant cause of global blindness. We used quantitative mass spectrometry to analyze the composition of XFM in lens capsule specimens and in aqueous humor (AH) samples from patients with XFS, patients with XFG and unaffected individuals. Methods: Pieces of lens capsule and samples of AH were obtained with consent from patients undergoing cataract surgery. Tryptic digests of capsule or AH were analyzed by high-performance liquid chromatography-mass spectrometry and relative differences between samples were quantified using the tandem mass tag technique. The distribution of XFM on the capsular surface was visualized by SEM and super-resolution light microscopy. Results: A small set of proteins was consistently upregulated in capsule samples from patients with XFS and patients with XFG, including microfibril components fibrillin-1, latent transforming growth factor-ß-binding protein-2 and latent transforming growth factor-ß-binding protein-3. Lysyl oxidase-like 1, a cross-linking enzyme associated with XFS in genetic studies, was an abundant XFM constituent. Ligands of the transforming growth factor-ß superfamily were prominent, including LEFTY2, a protein best known for its role in establishing the embryonic body axis. Elevated levels of LEFTY2 were also detected in AH from patients with XFG, a finding confirmed subsequently by ELISA. Conclusions: This analysis verified the presence of suspected XFM proteins and identified novel components. Quantitative comparisons between patient samples revealed a consistent XFM proteome characterized by strong expression of fibrillin-1, lysyl oxidase-like-1, and LEFTY2. Elevated levels of LEFTY2 in the AH of patients with XFG may serve as a biomarker for the disease.


Asunto(s)
Humor Acuoso/metabolismo , Cristalinas/metabolismo , Síndrome de Exfoliación/metabolismo , Glaucoma de Ángulo Abierto/metabolismo , Cápsula del Cristalino/metabolismo , Agregado de Proteínas/fisiología , Anciano , Anciano de 80 o más Años , Aminoácido Oxidorreductasas/metabolismo , Cromatografía Líquida de Alta Presión , Cristalinas/ultraestructura , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibrilina-1/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Proteínas de Unión a TGF-beta Latente/metabolismo , Factores de Determinación Derecha-Izquierda/metabolismo , Cápsula del Cristalino/ultraestructura , Masculino , Espectrometría de Masas , Microscopía Electrónica de Rastreo , Persona de Mediana Edad
6.
Biomaterials ; 278: 121133, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34571434

RESUMEN

The generation of mature ventricular cardiomyocytes (CMs) resembling adult CMs from human pluripotent stem cells (hPSCs) is necessary for disease modeling and drug discovery. To investigate the effect of self-organizing capacity on the generation of mature cardiac organoids (COs), we generated cardiac mesoderm cell-derived COs (CMC-COs) and CM-derived COs (CM-COs) and evaluated COs. CMC-COs exhibited more organized sarcomere structures and mitochondria, well-arranged t-tubule structures, and evenly distributed intercalated discs. Increased expressions of ventricular CM, cardiac metabolic, t-tubule formation, K+ ion channel, and junctional markers were confirmed in CMC-COs. Mature ventricular-like function such as faster motion vector speed, decreased beats per min, increased peak-to-peak duration, and prolonged APD50 and APD90 were observed in CMC-COs. Transcriptional profiling revealed that extracellular matrix-integrin, focal adhesion, and LEFTY-PITX2 signaling pathways are upregulated in CMC-COs. LEFTY knockdown affected ECM-integrin-FA signaling pathways in CMC-COs. Here, we found that high self-organizing capacity of CMCs is critical for the generation of mature and ventricular COs. We also demonstrated that LEFTY-PITX2 signaling plays key roles for CM maturation and specification into ventricular-like CM subtype in CMC-COs. CMC-COs are an attractive resource for disease modeling and drug discovery.


Asunto(s)
Proteínas de Homeodominio , Células Madre Pluripotentes Inducidas , Factores de Determinación Derecha-Izquierda , Miocitos Cardíacos , Células Madre Pluripotentes , Factores de Transcripción , Diferenciación Celular , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Determinación Derecha-Izquierda/metabolismo , Mesodermo , Organoides , Transducción de Señal , Factores de Transcripción/metabolismo , Proteína del Homeodomínio PITX2
7.
J Cardiovasc Transl Res ; 14(4): 636-646, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33409963

RESUMEN

Transforming growth factor-ß1 signaling pathways are known to involve in the development of post-infarction fibrosis, a process characterized by the aberrant activation, proliferation, and differentiation of fibroblasts, as well as the unbalanced turnover of extracellular matrix proteins. Recent studies have shown that Lefty1, a novel member of TGF-ß superfamily, acts as a brake on the TGF-ß signaling pathway in non-cardiac tissues. However, its role in myocardial infarction (MI)-induced fibrosis and left ventricular remodeling has not been fully elucidated. Here, for the first time, we reported that Lefty1 alleviated post-MI fibroblast proliferation, differentiation, and secretion through suppressing p-Smad2 and p-ERK1/2 signaling pathways in vivo and in vitro. In MI mice or TGF-ß1-treated neonatal rat cardiac fibroblasts (CFBs), the expression of Lefty1 was upregulated. Adenovirus-mediated overexpression of Lefty1 significantly attenuated TGF-ß1-induced CFBs' proliferation, differentiation, and collagen production. Using the adeno-associated virus approach, we confirmed that Lefty1 attenuates MI-induced cardiac injury, as evidenced by the decreased infarct size and preserved cardiac function. These results highlight the importance of Lefty1 in the prevention of post-MI fibrosis and may help identify potential targets for therapeutic intervention of cardiac fibrosis. Graphical abstract.


Asunto(s)
Factores de Determinación Derecha-Izquierda/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Proteína Smad2/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Fibrosis , Vectores Genéticos , Factores de Determinación Derecha-Izquierda/genética , Masculino , Ratones Endogámicos C57BL , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Fosforilación , Ratas Sprague-Dawley , Transducción de Señal , Factor de Crecimiento Transformador beta1/farmacología , Función Ventricular Izquierda
8.
Semin Cell Dev Biol ; 110: 11-18, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32571625

RESUMEN

The initial breaking of left-right (L-R) symmetry in the embryo is controlled by a motile-cilia-driven leftward fluid flow in the left-right organiser (LRO), resulting in L-R asymmetric gene expression flanking the LRO. Ultimately this results in left- but not right-sided activation of the Nodal-Pitx2 pathway in more lateral tissues. While aspects of the initial breaking event clearly vary between vertebrates, events in the Lateral Plate Mesoderm (LPM) are conserved through the vertebrate lineage. Evidence from model systems and humans highlights the role of cilia both in the initial symmetry breaking and in the ability of more lateral tissues to exhibit asymmetric gene expression. In this review we concentrate on the process of L-R determination in mouse and humans.


Asunto(s)
Tipificación del Cuerpo/genética , Cilios/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mecanotransducción Celular/genética , Mesodermo/metabolismo , Animales , Cilios/ultraestructura , Embrión de Mamíferos , Retroalimentación Fisiológica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Determinación Derecha-Izquierda/genética , Factores de Determinación Derecha-Izquierda/metabolismo , Mesodermo/crecimiento & desarrollo , Mesodermo/ultraestructura , Ratones , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Wnt3/genética , Proteína Wnt3/metabolismo , Proteína del Homeodomínio PITX2
9.
Dev Biol ; 470: 21-36, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33197427

RESUMEN

Nodal signaling is essential for mesoderm and endoderm formation, as well as neural plate induction and establishment of left-right asymmetry. However, the mechanisms controlling expression of Nodal pathway genes in these contexts are not fully known. Previously, we showed that Cdx1b induces expression of downstream Nodal signaling factors during early endoderm formation. In this study, we show that Cdx1b also regulates epithalamic asymmetry in zebrafish embryos by modulating expression of ndr2 and lft1. We first knocked down cdx1b with translation-blocking and splicing-blocking morpholinos (MOs). Most embryos injected with translation-blocking MOs showed absent ndr2, lft1 and pitx2c expression in the left dorsal diencephalon during segmentation and pharyngula stages accompanied by aberrant parapineal migration and habenular laterality at 72 â€‹h post fertilization (hpf). These defects were less frequent in embryos injected with splicing-blocking MO. To confirm the morphant phenotype, we next generated both zygotic (Z)cdx1b-/- and maternal zygotic (MZ)cdx1b-/- mutants by CRISPR-Cas9 mutagenesis. Expression of ndr2, lft1 and pitx2c was absent in the left dorsal diencephalon of a high proportion of MZcdx1b-/- mutants; however, aberrant dorsal diencephalic pitx2c expression patterns were observed at low frequency in Zcdx1b-/- mutant embryos. Correspondingly, dysregulated parapineal migration and habenular laterality were also observed in MZcdx1b-/- mutant embryos at 72 hpf. On the other hand, Kupffer's vesicle cilia length and number, expression pattern of spaw in the lateral plate mesoderm and pitx2c in the gut as well as left-right patterning of various visceral organs were not altered in MZcdx1b-/- mutants compared to wild-type embryos. Chromatin immunoprecipitation revealed that Cdx1b directly regulates ndr2 and lft1 expression. Furthermore, injection of cdx1b-vivo MO1 but not cdx1b-vivo 4 â€‹mm MO1 in the forebrain ventricle at 18 hpf significantly downregulated lft1 expression in the left dorsal diencephalon at 23-24 â€‹s stages. Together, our results suggest that Cdx1b regulates transcription of ndr2 and lft1 to maintain proper Nodal activity in the dorsal diencephalon and epithalamic asymmetry in zebrafish embryos.


Asunto(s)
Tipificación del Cuerpo/genética , Epitálamo/embriología , Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Factores de Determinación Derecha-Izquierda/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Animales , Movimiento Celular , Diencéfalo/embriología , Diencéfalo/metabolismo , Embrión no Mamífero/metabolismo , Epitálamo/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Habénula/embriología , Corazón/embriología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factores de Determinación Derecha-Izquierda/metabolismo , Proteína Nodal/metabolismo , Glándula Pineal/citología , Glándula Pineal/embriología , Unión Proteica , Transducción de Señal , Pez Cebra/metabolismo
10.
Mol Carcinog ; 59(12): 1409-1419, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33111989

RESUMEN

Glioblastomas (GBM) contain numerous hypoxic foci associated with a rare fraction of glioma stem cells (GSCs). Left-right determination factor (LEFTY) and Nodal, members of the transforming growth factor ß (TGF-ß) superfamily, have glycogen synthase kinase 3ß (GSK-3ß) phosphorylation motifs and are linked with stemness in human malignancies. Herein, we investigated the roles of LEFTY and Nodal in GBM hypoxic foci. In clinical samples, significantly higher expression of LEFTY, Nodal, phospho (p) GSK-3ß, pSmad2, and Nestin, as well as higher apoptotic and lower proliferation rates, were observed in nonpseudopalisading (non-Ps) perinecrotic lesions as compared to Ps and non-necrotic tumor lesions, with a positive correlation between LEFTY, Nodal, pGSK-3ß, or pSmad2 scores. In KS-1, a GBM cell line that lacks endogenous Nodal expression, treatment with the hypoxic mimetic CoCl2 increased LEFTY, pGSK-3ß, and pSmad2 levels, but decreased pAkt levels. Moreover, the promoter for LEFTY, but not Nodal, was activated by Smad2 or TGF-ß1, suggesting that overexpression of LEFTY and Nodal may be due to Akt-independent GSK-3ß inactivation, with or without cooperation of the TGF-ß1/Smad2 axis. LEFTY and Nodal overexpression increased proliferation rates and reduced susceptibility to CoCl2 -induced apoptosis, and increased the expression of epithelial-mesenchymal transition (EMT)/GSC-related markers. An increased ALDH1high population and more efficient spheroid formation was also observed in LEFTY-overexpressing cells. These findings suggest that LEFTY and Nodal may contribute to cell survival in non-Ps GBM perinecrotic lesions, leading to alterations in apoptosis, proliferation, or EMT/GCS features.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Factores de Determinación Derecha-Izquierda/metabolismo , Proteína Nodal/metabolismo , Regulación hacia Arriba , Adolescente , Adulto , Anciano , Neoplasias Encefálicas/genética , Hipoxia de la Célula , Línea Celular Tumoral , Supervivencia Celular , Cobalto/efectos adversos , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Humanos , Factores de Determinación Derecha-Izquierda/genética , Masculino , Persona de Mediana Edad , Proteína Nodal/genética , Fosforilación , Transducción de Señal , Adulto Joven
11.
Mol Immunol ; 126: 31-39, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32745796

RESUMEN

Activated hepatic stellate cells (HSCs) are the major cell type involved in the deposition of extracellular matrix (ECM) during the development of hepatic fibrosis. In this study, we revealed that left-right determination factor 2 (LEFTY2), one of the proteins belonging to the transforming growth factor-ß (TGF-ß) protein superfamily, was remarkedly decreased in human hepatic fibrosis tissues and in a carbon tetrachloride (CCl4)-induced liver fibrosis mouse model. In addition, TGF-ß1 treatment markedly reduced the level of LEFTY2 in HSCs. Importantly, overexpression of LEFTY2 suppressed the activation and proliferation of HSCs. LEFTY2 inhibited the expression of TGF-ß1-induced fibrosis-associated genes (α-SMA and COL1a1) in human (LX-2) and rat (HSC-T6) HSC cell lines in vitro. Mechanistically, we demonstrated, for the first time, the role of LEFTY2 in inhibiting TGF-ß1/Smad3 signaling, suggesting that there is a mutual antagonism between LEFTY2 and TGF-ß1/Smad3 signaling during liver fibrosis. Similarly, we observed that LEFTY2 has a negative effect on its downstream genes, including c-MYC, CDK4, and cyclin D1, in liver fibrosis. Collectively, our data strongly indicated that LEFTY2 plays an important role in controlling the proliferation and activation of HSCs in the progression of liver fibrosis and this could be a potential therapeutic target for its treatment.


Asunto(s)
Células Estrelladas Hepáticas/patología , Factores de Determinación Derecha-Izquierda/metabolismo , Cirrosis Hepática/patología , Hígado/patología , Anciano , Animales , Tetracloruro de Carbono/toxicidad , Línea Celular , Proliferación Celular , Regulación hacia Abajo , Femenino , Humanos , Hígado/efectos de los fármacos , Cirrosis Hepática/inducido químicamente , Masculino , Ratones , Persona de Mediana Edad , Ratas , Transducción de Señal , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
12.
J Mol Histol ; 51(1): 77-87, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32065356

RESUMEN

Epithelial-mesenchymal transition (EMT) is a biological process in which tubular epithelial cells lose their phenotypes, and new mesenchymal feature are obtained. In particular, type II EMT possibly contributes to renal tissue fibrogenesis. Recent studies indicate that Lefty-1, a novel member of the TGF-ß superfamily with pleiotropical and biological regulation characteristics on TGF-ß and other signaling pathways, is considered to have potential fibrotic effects. However, its role in EMT, which is often a long-term consequence of renal tubulointerstitial fibrosis, remains unknown. In this study, we found that Lefty-1 alleviates EMT induction through antagonizing TGF-ß/Smad pathway in vivo and in vitro. In unilateral ureteral obstruction (UUO) model mice, administration of adenovirus-mediated overexpression of Lefty-1 (Ad-Lefty-1) significantly reduced TGF-ß1/Smad expression and alleviated the phenotypic transition of epithelial cells to mesenchymal cells and extracellular matrix (ECM) accumulation. In high glucose-induced rat renal tubular duct epithelial cell line (NRK-52E), EMT and ECM synthesis were alleviated with Lefty-1 treatment, which significantly inhibited TGF-ß1/Smad pathway activation in UUO mice and high glucose-treated NRK-52E cells. Thus, Lefty-1 can alleviate EMT and renal interstitial fibrosis in vivo and in vitro through antagonizing the TGF-ß/Smad pathway, and Lefty-1 might have a potential novel therapeutic effect on fibrotic kidney diseases.


Asunto(s)
Transición Epitelial-Mesenquimal , Túbulos Renales/metabolismo , Factores de Determinación Derecha-Izquierda/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/metabolismo , Animales , Línea Celular , Fibrosis , Túbulos Renales/patología , Masculino , Ratones , Obstrucción Ureteral/patología
13.
PLoS Biol ; 17(8): e3000203, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31430272

RESUMEN

Zebrafish dorsal forerunner cells (DFCs) undergo vigorous proliferation during epiboly and then exit the cell cycle to generate Kupffer's vesicle (KV), a ciliated organ necessary for establishing left-right (L-R) asymmetry. DFC proliferation defects are often accompanied by impaired cilia elongation in KV, but the functional and molecular interaction between cell-cycle progression and cilia formation remains unknown. Here, we show that chemokine receptor Cxcr4a is required for L-R laterality by controlling DFC proliferation and KV ciliogenesis. Functional analysis revealed that Cxcr4a accelerates G1/S transition in DFCs and stabilizes forkhead box j1a (Foxj1a), a master regulator of motile cilia, by stimulating Cyclin D1 expression through extracellular regulated MAP kinase (ERK) 1/2 signaling. Mechanistically, Cyclin D1-cyclin-dependent kinase (CDK) 4/6 drives G1/S transition during DFC proliferation and phosphorylates Foxj1a, thereby disrupting its association with proteasome 26S subunit, non-ATPase 4b (Psmd4b), a 19S regulatory subunit. This prevents the ubiquitin (Ub)-independent proteasomal degradation of Foxj1a. Our study uncovers a role for Cxcr4 signaling in L-R patterning and provides fundamental insights into the molecular linkage between cell-cycle progression and ciliogenesis.


Asunto(s)
Cilios/metabolismo , Factores de Determinación Derecha-Izquierda/metabolismo , Receptores CXCR4/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Tipificación del Cuerpo/genética , Ciclo Celular/fisiología , División Celular , Proliferación Celular , Quimiocinas/metabolismo , Embrión no Mamífero/metabolismo , Factores de Transcripción Forkhead/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Morfogénesis , Receptores CXCR4/fisiología , Transducción de Señal , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
14.
Dev Biol ; 456(1): 63-73, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31419410

RESUMEN

The organizer is an essential signaling center required for axial formation during vertebrate embryonic development. In the basal chordate amphioxus, the dorsal blastopore lip of the gastrula has been proposed to be homologous to the vertebrate organizer. Lefty is one of the first genes to be expressed in the organizer. The present results show that Lefty overexpression abolishes the organizer; the embryos were severely ventralized and posteriorized, and failed to develop anterior and dorsal structures. In Lefty knockouts the organizer is enlarged, and anterior and dorsal structures are expanded. Different from Lefty morphants in vertebrates, amphioxus Lefty mutants also exhibited left-right defects. Inhibition of Nodal with SB505124 partially rescued the effects of Lefty loss-of-function on morphology. In addition, while SB505124 treatment blocked Lefty expression in the cleavage stages of amphioxus embryos, activation of Nodal signaling with Activin protein induced ectopic Lefty expression at these stages. These results show that the interplay between Lefty and Nodal signaling plays an essential role in the specification of the amphioxus organizer and axes.


Asunto(s)
Anfioxos/embriología , Factores de Determinación Derecha-Izquierda/metabolismo , Proteína Nodal/metabolismo , Activinas/metabolismo , Animales , Tipificación del Cuerpo/genética , Femenino , Gástrula/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Anfioxos/metabolismo , Factores de Determinación Derecha-Izquierda/fisiología , Masculino , Proteína Nodal/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo
15.
Biosystems ; 173: 281-297, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30292532

RESUMEN

The expression of the TGF-ß protein Nodal on the left side of vertebrate embryos is a determining event in the development of internal-organ asymmetry. We present a mathematical model for the control of the expression of Nodal and its antagonist Lefty consisting entirely of realistic elementary reactions. We analyze the model in the absence of Lefty and find a wide range of parameters over which bistability (two stable steady states) is observed, with one stable steady state a low-Nodal state corresponding to the right-hand developmental fate, and the other a high-Nodal state corresponding to the left. We find that bistability requires a transcription factor containing two molecules of phosphorylated Smad2. A numerical survey of the full model, including Lefty, shows the effects of Lefty on the potential for bistability, and on the conditions that lead to the system reaching one or the other steady state.


Asunto(s)
Tipificación del Cuerpo , Factores de Determinación Derecha-Izquierda/metabolismo , Modelos Biológicos , Proteína Nodal/metabolismo , Proteína Smad2/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Mamíferos , Ratones , Fosforilación , Transducción de Señal , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo
16.
Nat Commun ; 9(1): 4261, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30323168

RESUMEN

Barrett's oesophagus is a precursor of oesophageal adenocarcinoma. In this common condition, squamous epithelium in the oesophagus is replaced by columnar epithelium in response to acid reflux. Barrett's oesophagus is highly heterogeneous and its relationships to normal tissues are unclear. Here we investigate the cellular complexity of Barrett's oesophagus and the upper gastrointestinal tract using RNA-sequencing of single cells from multiple biopsies from six patients with Barrett's oesophagus and two patients without oesophageal pathology. We find that cell populations in Barrett's oesophagus, marked by LEFTY1 and OLFM4, exhibit a profound transcriptional overlap with oesophageal submucosal gland cells, but not with gastric or duodenal cells. Additionally, SPINK4 and ITLN1 mark cells that precede morphologically identifiable goblet cells in colon and Barrett's oesophagus, potentially aiding the identification of metaplasia. Our findings reveal striking transcriptional relationships between normal tissue populations and cells in a premalignant condition, with implications for clinical practice.


Asunto(s)
Esófago de Barrett/genética , Epitelio/patología , Esófago/patología , Análisis de Secuencia de ARN , Análisis de la Célula Individual/métodos , Transcripción Genética , Esófago de Barrett/patología , Células Caliciformes/metabolismo , Células Caliciformes/patología , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Factores de Determinación Derecha-Izquierda/metabolismo , ARN Mensajero/metabolismo , Regulación hacia Arriba
17.
Nat Cell Biol ; 20(9): 1032-1042, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30061678

RESUMEN

Individuals can vary substantially in size, but the proportions of their body plans are often maintained. We generated smaller zebrafish by removing 30% of their cells at the blastula stages and found that these embryos developed into normally patterned individuals. Strikingly, the proportions of all germ layers adjusted to the new embryo size within 2 hours after cell removal. As Nodal-Lefty signalling controls germ-layer patterning, we performed a computational screen for scale-invariant models of this activator-inhibitor system. This analysis predicted that the concentration of the highly diffusive inhibitor Lefty increases in smaller embryos, leading to a decreased Nodal activity range and contracted germ-layer dimensions. In vivo studies confirmed that Lefty concentration increased in smaller embryos, and embryos with reduced Lefty levels or with diffusion-hindered Lefty failed to scale their tissue proportions. These results reveal that size-dependent inhibition of Nodal signalling allows scale-invariant patterning.


Asunto(s)
Blástula/metabolismo , Tipificación del Cuerpo , Factores de Determinación Derecha-Izquierda/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/genética , Regulación del Desarrollo de la Expresión Génica , Factores de Determinación Derecha-Izquierda/genética , Proteínas de la Membrana/genética , Transducción de Señal , Factores de Tiempo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
18.
Int J Mol Med ; 42(3): 1229-1236, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29845221

RESUMEN

Endometrial cancer (EC) is the most common gynecological tumor in developed countries with an increasing incidence. Left­right determination factor 2 (LEFTY2), a suppressor of cell proliferation and tumor growth, is a negative regulator of EC progression. The roles of LEFTY2 are emerging; however, the regulatory mechanisms of its expression have not been well understood. MicroRNA (miR)­215 as an oncogene serves an important role in tumorigenesis by regulating target genes. In the present study, it was demonstrated that overexpression of miR­215 promoted epithelial to mesenchymal transition (EMT), colony formation and DNA synthesis in EC HEC­1A cells and its expression was upregulated in EC tissues. Using online miR target prediction software, it was revealed that LEFTY2 is predicted as a target of miR­215. Using western blot analysis and immunofluorescence assays, it was demonstrated that overexpression of miR­215 markedly downregulated LEFTY2 protein expression levels in HEC­1A cells and LEFTY2 protein expression was downregulated in EC tissues, which was inversely correlated with miR­215 expression. Furthermore, the present study indicated that overexpression of LEFTY2 protein promoted mesenchymal to epithelial transition and sensitized HEC­1A cells to cisplatin treatment. In addition, it was revealed that the overexpression of LEFTY2 inhibited colony formation and DNA synthesis in HEC­1A cells. Thus, miR­215 may promote EMT and proliferation by regulating LEFTY2 in EC.


Asunto(s)
Neoplasias Endometriales/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Factores de Determinación Derecha-Izquierda/metabolismo , MicroARNs/metabolismo , Adulto , Anciano , Western Blotting , Proliferación Celular/genética , Proliferación Celular/fisiología , Biología Computacional , Neoplasias Endometriales/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Factores de Determinación Derecha-Izquierda/genética , MicroARNs/genética , Persona de Mediana Edad
19.
Nat Commun ; 9(1): 1942, 2018 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-29769531

RESUMEN

The establishment of left-right (LR) asymmetry is fundamental to animal development, but the identification of a unifying mechanism establishing laterality across different phyla has remained elusive. A cilia-driven, directional fluid flow is important for symmetry breaking in numerous vertebrates, including zebrafish. Alternatively, LR asymmetry can be established independently of cilia, notably through the intrinsic chirality of the acto-myosin cytoskeleton. Here, we show that Myosin1D (Myo1D), a previously identified regulator of Drosophila LR asymmetry, is essential for the formation and function of the zebrafish LR organizer (LRO), Kupffer's vesicle (KV). Myo1D controls the orientation of LRO cilia and interacts functionally with the planar cell polarity (PCP) pathway component VanGogh-like2 (Vangl2), to shape a productive LRO flow. Our findings identify Myo1D as an evolutionarily conserved regulator of animal LR asymmetry, and show that functional interactions between Myo1D and PCP are central to the establishment of animal LR asymmetry.


Asunto(s)
Tipificación del Cuerpo/genética , Miosinas/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Polaridad Celular/genética , Cilios/genética , Cilios/metabolismo , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Determinación Derecha-Izquierda/genética , Factores de Determinación Derecha-Izquierda/metabolismo , Mutación , Miosinas/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
20.
Nucleic Acids Res ; 46(12): 6026-6040, 2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29733394

RESUMEN

Sin3a is a core component of histone-deacetylation-activity-associated transcriptional repressor complex, playing important roles in early embryo development. Here, we reported that down-regulation of Sin3a led to the loss of embryonic stem cell (ESC) self-renewal and skewed differentiation into mesendoderm lineage. We found that Sin3a functioned as a transcriptional coactivator of the critical Nodal antagonist Lefty1 through interacting with Tet1 to de-methylate the Lefty1 promoter. Further studies showed that two amino acid residues (Phe147, Phe182) in the PAH1 domain of Sin3a are essential for Sin3a-Tet1 interaction and its activity in regulating pluripotency. Furthermore, genome-wide analyses of Sin3a, Tet1 and Pol II ChIP-seq and of 5mC MeDIP-seq revealed that Sin3a acted with Tet1 to facilitate the transcription of a set of their co-target genes. These results link Sin3a to epigenetic DNA modifications in transcriptional activation and have implications for understanding mechanisms underlying versatile functions of Sin3a in mouse ESCs.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Activación Transcripcional , Animales , Diferenciación Celular , Línea Celular , Linaje de la Célula , Células Madre Embrionarias/citología , Factores de Determinación Derecha-Izquierda/genética , Factores de Determinación Derecha-Izquierda/metabolismo , Ratones , Proteína Nodal/metabolismo , Regiones Promotoras Genéticas , Dominios y Motivos de Interacción de Proteínas , Proteínas Represoras/química , Proteínas Represoras/genética , Complejo Correpresor Histona Desacetilasa y Sin3
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA