Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 7(1): 527, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714733

RESUMEN

Macrophages are versatile cells of the innate immune system that work by altering their pro- or anti-inflammatory features. Their dysregulation leads to inflammatory disorders such as inflammatory bowel disease. We show that macrophage-specific upregulation of the clock output gene and transcription factor E4BP4 reduces the severity of colitis in mice. RNA-sequencing and single-cell analyses of macrophages revealed that increased expression of E4BP4 leads to an overall increase in expression of anti-inflammatory genes including Il4ra with a concomitant reduction in pro-inflammatory gene expression. In contrast, knockout of E4BP4 in macrophages leads to increased proinflammatory gene expression and decreased expression of anti-inflammatory genes. ChIP-seq and ATAC-seq analyses further identified Il4ra as a target of E4BP4, which drives anti-inflammatory polarization in macrophages. Together, these results reveal a critical role for E4BP4 in regulating macrophage inflammatory phenotypes and resolving inflammatory bowel diseases.


Asunto(s)
Colitis , Macrófagos , Animales , Macrófagos/inmunología , Macrófagos/metabolismo , Colitis/genética , Colitis/inmunología , Colitis/metabolismo , Colitis/patología , Colitis/inducido químicamente , Ratones , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Ratones Noqueados , Fenotipo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Índice de Severidad de la Enfermedad , Masculino , Inflamación/genética , Inflamación/metabolismo
2.
Front Genet ; 15: 1383176, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38601074

RESUMEN

Introduction: RRAS2, a member of the R-Ras subfamily of Ras-like low-molecular-weight GTPases, is considered to regulate cell proliferation and differentiation via the RAS/MAPK signaling pathway. Seven RRAS2 pathogenic variants have been reported in patients with Noonan syndrome; however, few functional analyses have been conducted. Herein, we report two patients who presented with a Noonan-like phenotype with recurrent and novel RRAS2 pathogenic variants (p.Gly23Val and p.Gly24Glu, respectively) and the results of their functional analysis. Materials and methods: Wild-type (WT) and mutant RRAS2 genes were transiently expressed in Human Embryonic Kidney293 cells. Expression of RRAS2 and phosphorylation of ERK1/2 were confirmed by Western blotting, and the RAS signaling pathway activity was measured using a reporter assay system with the serum response element-luciferase construct. WT and p.Gly23Val RRAS2 were expressed in Drosophila eye using the glass multiple reporter-Gal4 driver. Mutant mRNA microinjection into zebrafish embryos was performed, and the embryo jaws were observed. Results: No obvious differences in the expression of proteins WT, p.Gly23Val, and p.Gly24Glu were observed. The luciferase reporter assay showed that the activity of p.Gly23Val was 2.45 ± 0.95-fold higher than WT, and p.Gly24Glu was 3.06 ± 1.35-fold higher than WT. For transgenic flies, the p.Gly23Val expression resulted in no adults flies emerging, indicating lethality. For mutant mRNA-injected zebrafish embryos, an oval shape and delayed jaw development were observed compared with WT mRNA-injected embryos. These indicated hyperactivity of the RAS signaling pathway. Discussion: Recurrent and novel RRAS2 variants that we reported showed increased in vitro or in vivo RAS signaling pathway activity because of gain-of-function RRAS2 variants. Clinical features are similar to those previously reported, suggesting that RRAS2 gain-of-function variants cause this disease in patients.

3.
PLoS One ; 17(10): e0269077, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36194562

RESUMEN

Ewing's sarcoma is the second most common bone malignancy in children or young adults and is caused by an oncogenic transcription factor by a chromosomal translocation between the EWSR1 gene and the ETS transcription factor family. However, the transcriptional mechanism of EWS-ETS fusion proteins is still unclear. To identify the transcriptional complexes of EWS-ETS fusion transcription factors, we applied a proximal labeling system called BioID in Ewing's sarcoma cells. We identified AHDC1 as a proximal protein of EWS-ETS fusion proteins. AHDC1 knockdown showed a reduced cell growth and transcriptional activity of EWS-FLI1. AHDC1 knockdown also reduced BRD4 and BRG1 protein levels, both known as interacting proteins of EWS-FLI1. Our results suggest that AHDC1 supports cell growth through EWS-FLI1.


Asunto(s)
Sarcoma de Ewing , Proteínas de Ciclo Celular/metabolismo , Niño , ADN , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/genética , Proteína EWS de Unión a ARN/metabolismo , Sarcoma de Ewing/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Commun Biol ; 2: 364, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31602413

RESUMEN

The pineal gland functioning as a photoreceptive organ in non-mammalian species is a serial homolog of the retina. Here we found that Brain-specific homeobox (Bsx) is a key regulator conferring individuality on the pineal gland between the two serially homologous photoreceptive organs in zebrafish. Bsx knock-down impaired the pineal development with reduced expression of exorh, the pineal-specific gene responsible for the photoreception, whereas it induced ectopic expression of rho, a retina-specific gene, in the pineal gland. Bsx remarkably transactivated the exorh promoter in combination with Otx5, but not with Crx, through its binding to distinct subtypes of PIRE, a DNA cis-element driving Crx/Otx-dependent pineal-specific gene expression. These results demonstrate that the identity of pineal photoreceptive neurons is determined by the combinatorial code of Bsx and Otx5, the former confers the pineal specificity at the tissue level and the latter determines the photoreceptor specificity at the cellular level.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Glándula Pineal/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Factores de Transcripción Otx/metabolismo , Factor de Transcripción PAX6/metabolismo , Glándula Pineal/citología , Glándula Pineal/crecimiento & desarrollo , Regiones Promotoras Genéticas , Rodopsina/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Pez Cebra , Proteínas de Pez Cebra/genética
5.
Sci Rep ; 9(1): 196, 2019 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-30655599

RESUMEN

The circadian clock generates behavioral rhythms to maximize an organism's physiological efficiency. Light induces the formation of these rhythms by synchronizing cellular clocks. In zebrafish, the circadian clock components Period2 (zPER2) and Cryptochrome1a (zCRY1a) are light-inducible, however their physiological functions are unclear. Here, we investigated the roles of zPER2 and zCRY1a in regulating locomotor activity and behavioral rhythms. zPer2/zCry1a double knockout (DKO) zebrafish displayed defects in total locomotor activity and in forming behavioral rhythms when briefly exposed to light for 3-h. Exposing DKO zebrafish to 12-h light improved behavioral rhythm formation, but not total activity. Our data suggest that the light-inducible circadian clock regulator zCRY2a supports rhythmicity in DKO animals exposed to 12-h light. Single cell imaging analysis revealed that zPER2, zCRY1a, and zCRY2a function in synchronizing cellular clocks. Furthermore, microarray analysis of DKO zebrafish showed aberrant expression of genes involved regulating cellular metabolism, including ATP production. Overall, our results suggest that zPER2, zCRY1a and zCRY2a help to synchronize cellular clocks in a light-dependent manner, thus contributing to behavioral rhythm formation in zebrafish. Further, zPER2 and zCRY1a regulate total physical activity, likely via regulating cellular energy metabolism. Therefore, these circadian clock components regulate the rhythmicity and amount of locomotor behavior.


Asunto(s)
Relojes Circadianos/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Proteínas CLOCK/fisiología , Criptocromos/fisiología , Luz , Locomoción , Proteínas Circadianas Period/fisiología , Análisis de la Célula Individual , Proteínas de Pez Cebra/fisiología
6.
Methods Mol Biol ; 1893: 167-181, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30565134

RESUMEN

The transcription coactivator, Yes-associated protein (YAP), which is a nuclear effector of the Hippo signaling pathway, has been shown to be a mechano-transducer. By using mutant fish and human 3D spheroids, we have recently demonstrated that YAP is also a mechano-effector. YAP functions in three-dimensional (3D) morphogenesis of organ and global body shape by controlling actomyosin-mediated tissue tension. In this chapter, we present a platform that links the findings in fish embryos with human cells. The protocols for analyzing tissue tension-mediated global body shape/organ morphogenesis in vivo and ex vivo using medaka fish embryos and in vitro using human cell spheroids represent useful tools for unraveling the molecular mechanisms by which YAP functions in regulating global body/organ morphogenesis.


Asunto(s)
Desarrollo Embrionario/genética , Morfogénesis/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética , Animales , Técnicas de Cultivo de Célula , Proteínas de Ciclo Celular , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Humanos , Mutación , Proteínas Nucleares/metabolismo , Oryzias , Esferoides Celulares , Factores de Transcripción/metabolismo
7.
Curr Opin Cell Biol ; 49: 64-70, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29253723

RESUMEN

Cells of terrestrial animals are constantly exposed to external forces including gravity. However, the complex 3D structure of the body and its organs form without being flattened. A century ago, the mathematical biologist D'Arcy Thompson predicted in 'On Growth and Form' that terrestrial animal body shapes are entirely conditioned by gravity [1], but the prediction remained to be proved due to the lack of an appropriate animal model. In this review, we outline a new mechanism of morphogenesis which ensures the generation of vertebrate 3D body shape that can withstand gravity and in which Hippo-YAP signaling acts as a mechano-effector controlling mechano-homeostasis. We will highlight the recent papers that advanced the field and discuss the impact of this previously unrecognized function of YAP-mediated signaling on the established concept of organogenesis, tissue homeostasis and disease.


Asunto(s)
Homeostasis/genética , Modelos Biológicos , Proteínas Nucleares/genética , Organogénesis/genética , Factores de Transcripción/genética , Animales , Proteínas de Ciclo Celular , Humanos , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo
8.
Dev Growth Differ ; 59(1): 52-58, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28093734

RESUMEN

Cells of our body are constantly exposed to physical forces such as tissue tension. In recent years, it has been shown that such mechanical signals greatly influence a number of cellular processes, including proliferation, differentiation, and migration. Conversely, cells maintain the mechanical properties of tissues by remodeling their own extracellular environment. To date, however, it is unclear about the molecular mechanisms to maintain the mechanical environment ("mechano-homeostasis") in which extracellular mechanical cues are integrated with cell proliferation and differentiation to ensure tissue, organ and body form. In this review, we outline the molecular basis of mechanotransduction, and overview some useful techniques for measuring cellular tension. In the latter part, we describe our recent finding that a transcriptional cofactor YAP plays a crucial role in three-dimensional organ formation and its maintenance by controlling tissue tension, and functions as a key molecule governing mechano-homeostasis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Embrión de Mamíferos/embriología , Mecanotransducción Celular/fisiología , Organogénesis/fisiología , Fosfoproteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Embrión de Mamíferos/citología , Humanos , Fosfoproteínas/genética , Factores de Transcripción , Proteínas Señalizadoras YAP
9.
Sci Rep ; 6: 37697, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27883036

RESUMEN

The primitive streak in peri-implantation embryos forms the mesoderm and endoderm and controls cell differentiation. The metabolic cues regulating primitive streak formation remain largely unknown. Here we utilised a mouse embryonic stem (ES) cell differentiation system and a library of well-characterised drugs to identify these metabolic factors. We found that statins, which inhibit the mevalonate metabolic pathway, suppressed primitive streak formation in vitro and in vivo. Using metabolomics and pharmacologic approaches we identified the downstream signalling pathway of mevalonate and revealed that primitive streak formation requires protein farnesylation but not cholesterol synthesis. A tagging-via-substrate approach revealed that nuclear lamin B1 and small G proteins were farnesylated in embryoid bodies and important for primitive streak gene expression. In conclusion, protein farnesylation driven by the mevalonate pathway is a metabolic cue essential for primitive streak formation.


Asunto(s)
Redes y Vías Metabólicas , Ácido Mevalónico/metabolismo , Línea Primitiva/embriología , Línea Primitiva/metabolismo , Prenilación de Proteína , Animales , Diferenciación Celular , Regulación hacia Abajo/genética , Cuerpos Embrioides , Regulación del Desarrollo de la Expresión Génica , Metaboloma , Metabolómica , Ratones Endogámicos ICR , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Neurogénesis , Análisis de Secuencia por Matrices de Oligonucleótidos , Organogénesis , Pez Cebra
10.
Biochem Biophys Res Commun ; 474(1): 146-153, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-27105919

RESUMEN

The precise government of the left-right (LR) specification of an organ is an essential aspect of its morphogenesis. Multiple signaling cascades have been implicated in the establishment of vertebrate LR asymmetry. Recently, mTOR signaling was found to critically regulate the development of LR asymmetry in zebrafish. However, the upstream factor(s) that activate mTOR signaling in the context of LR specification are as yet unknown. In this study, we identify the SLC7 amino acid transporters Slc7a7 and Slc7a8 as novel regulators of LR asymmetry development in the small fish medaka. Knockdown of Slc7a7 and/or Slc7a8 in medaka embryos disrupted LR organ asymmetries. Depletion of Slc7a7 hindered left-sided expression of the southpaw (spaw) gene, which is responsible for LR axis determination. Work at the cellular level revealed that Slc7a7 coordinates ciliogenesis in the epithelium of Kupffer's vesicle and thereby the generation of the nodal fluid flow required for LR asymmetry. Interestingly, knockdown of Slc7a7 depressed mTOR signaling activity in medaka embryos. Treatment with rapamycin, an inhibitor of mTOR signaling, together with Slc7a7 knockdown synergistically perturbed spaw expression, indicating an interaction between Slc7a7 and mTOR signaling affecting gene expression required for LR specification. Taken together, our results demonstrate that Slc7a7 governs the regulation of LR asymmetry development via the activation of mTOR signaling.


Asunto(s)
Tipificación del Cuerpo/fisiología , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/metabolismo , Organogénesis/fisiología , Oryzias/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Sistema de Transporte de Aminoácidos y+L , Animales , Regulación del Desarrollo de la Expresión Génica/fisiología , Transducción de Señal/fisiología
11.
Anal Chem ; 88(1): 838-44, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26597767

RESUMEN

Apoptosis plays a pivotal role in development and tissue homeostasis in multicellular organisms. Dysfunction of apoptosis is involved in many fatal diseases such as cancer. Visualization of apoptosis in living animals is necessary to understand the mechanism of apoptosis-related diseases. Here, we describe a genetically encoded fluorescent probe for imaging apoptosis in living multicellular organisms, based on spontaneous complementation of two fragments of a green fluorescent protein (GFP) variant (GFP OPT). The probe is designed for detection of mitochondria-mediated apoptosis during which a mitochondrial protein of Smac is released into cytosol. The Smac is connected with a carboxy-terminal fragment of GFP OPT (GFP11), whereas the remainder of GFP OPT (GFP(1-10)) is located in the cytosol. Under an apoptotic condition, the Smac is released from mitochondria into cytosol, allowing complementation of the GFP-OPT fragments and the emission of fluorescence. Live-cell imaging demonstrates that the probe enables detection of apoptosis in living cells with a high signal-to-background ratio. We applied the probe to living zebrafish, in which apoptotic cells were visualized with fluorescence. The technique provides a useful tool for the study of apoptosis in living animals, facilitating elucidation of the mechanisms of apoptosis-related diseases.


Asunto(s)
Apoptosis/genética , Colorantes Fluorescentes/metabolismo , Prueba de Complementación Genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Animales , Supervivencia Celular , Células HeLa , Humanos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Células Tumorales Cultivadas , Pez Cebra
12.
Nature ; 521(7551): 217-221, 2015 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-25778702

RESUMEN

Vertebrates have a unique 3D body shape in which correct tissue and organ shape and alignment are essential for function. For example, vision requires the lens to be centred in the eye cup which must in turn be correctly positioned in the head. Tissue morphogenesis depends on force generation, force transmission through the tissue, and response of tissues and extracellular matrix to force. Although a century ago D'Arcy Thompson postulated that terrestrial animal body shapes are conditioned by gravity, there has been no animal model directly demonstrating how the aforementioned mechano-morphogenetic processes are coordinated to generate a body shape that withstands gravity. Here we report a unique medaka fish (Oryzias latipes) mutant, hirame (hir), which is sensitive to deformation by gravity. hir embryos display a markedly flattened body caused by mutation of YAP, a nuclear executor of Hippo signalling that regulates organ size. We show that actomyosin-mediated tissue tension is reduced in hir embryos, leading to tissue flattening and tissue misalignment, both of which contribute to body flattening. By analysing YAP function in 3D spheroids of human cells, we identify the Rho GTPase activating protein ARHGAP18 as an effector of YAP in controlling tissue tension. Together, these findings reveal a previously unrecognised function of YAP in regulating tissue shape and alignment required for proper 3D body shape. Understanding this morphogenetic function of YAP could facilitate the use of embryonic stem cells to generate complex organs requiring correct alignment of multiple tissues.


Asunto(s)
Tamaño Corporal/genética , Proteínas de Peces/metabolismo , Morfogénesis/genética , Oryzias/anatomía & histología , Oryzias/embriología , Actomiosina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Embrión no Mamífero/anatomía & histología , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Proteínas de Peces/genética , Proteínas Activadoras de GTPasa/metabolismo , Genes Esenciales/genética , Gravitación , Humanos , Mutación/genética , Tamaño de los Órganos/genética , Oryzias/genética , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Esferoides Celulares/citología , Esferoides Celulares/metabolismo
13.
Biochem Biophys Res Commun ; 457(4): 493-9, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25576873

RESUMEN

Mammalian ovarian G-protein-coupled receptor 1 (OGR1) and GPR4 are identified as a proton-sensing G-protein-coupled receptor coupling to multiple intracellular signaling pathways. In the present study, we examined whether zebra fish OGR1 and GPR4 homologs (zOGR1 and zGPR4) could sense protons and activate the multiple intracellular signaling pathways and, if so, whether the similar positions of histidine residue, which is critical for sensing protons in mammalian OGR and GPR4, also play a role to sense protons and activate the multiple signaling pathways in the zebra fish receptors. We found that extracellular acidic pH stimulated CRE-, SRE-, and NFAT-promoter activities in zOGR1 overexpressed cells and stimulated CRE- and SRE- but not NFAT-promoter activities in zGPR4 overexpressed cells. The substitution of histidine residues at the 12th, 15th, 162th, and 264th positions from the N-terminal of zOGR1 with phenylalanine attenuated the proton-induced SRE-promoter activities. The mutation of the histidine residue at the 78th but not the 84th position from the N-terminal of zGPR4 to phenylalanine attenuated the proton-induced SRE-promoter activities. These results suggest that zOGR1 and zGPR4 are also proton-sensing G-protein-coupled receptors, and the receptor activation mechanisms may be similar to those of the mammalian receptors.


Asunto(s)
Protones , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Regulación de la Expresión Génica , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Datos de Secuencia Molecular , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Alineación de Secuencia , Transducción de Señal , Pez Cebra/genética , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/genética
14.
PLoS One ; 9(5): e97365, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24828882

RESUMEN

The precise regulation of numbers and types of neurons through control of cell cycle exit and terminal differentiation is an essential aspect of neurogenesis. The Hippo signaling pathway has recently been identified as playing a crucial role in promoting cell cycle exit and terminal differentiation in multiple types of stem cells, including in retinal progenitor cells. When Hippo signaling is activated, the core Mst1/2 kinases activate the Lats1/2 kinases, which in turn phosphorylate and inhibit the transcriptional cofactor Yap. During mouse retinogenesis, overexpression of Yap prolongs progenitor cell proliferation, whereas inhibition of Yap decreases this proliferation and promotes retinal cell differentiation. However, to date, it remains unknown how the Hippo pathway affects the differentiation of distinct neuronal cell types such as photoreceptor cells. In this study, we investigated whether Hippo signaling regulates retinogenesis during early zebrafish development. Knockdown of zebrafish mst2 induced early embryonic defects, including altered retinal pigmentation and morphogenesis. Similar abnormal retinal phenotypes were observed in zebrafish embryos injected with a constitutively active form of yap [(yap (5SA)]. Loss of Yap's TEAD-binding domain, two WW domains, or transcription activation domain attenuated the retinal abnormalities induced by yap (5SA), indicating that all of these domains contribute to normal retinal development. Remarkably, yap (5SA)-expressing zebrafish embryos displayed decreased expression of transcription factors such as otx5 and crx, which orchestrate photoreceptor cell differentiation by activating the expression of rhodopsin and other photoreceptor cell genes. Co-immunoprecipitation experiments revealed that Rx1 is a novel interacting partner of Yap that regulates photoreceptor cell differentiation. Our results suggest that Yap suppresses the differentiation of photoreceptor cells from retinal progenitor cells by repressing Rx1-mediated transactivation of photoreceptor cell genes during zebrafish retinogenesis.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Células Fotorreceptoras/fisiología , Proteínas Serina-Treonina Quinasas/genética , Retina/fisiología , Células Madre/fisiología , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica/genética , Morfogénesis/genética , Morfogénesis/fisiología , Neurogénesis/genética , Neurogénesis/fisiología , Neuronas/fisiología , Fosfoproteínas/genética , Pigmentación/genética , Pigmentación/fisiología , Rodopsina , Serina-Treonina Quinasa 3 , Transducción de Señal/genética , Factores de Transcripción/genética , Pez Cebra/fisiología
15.
Dis Model Mech ; 6(4): 905-14, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23720231

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is a condition in which excessive fat accumulates in the liver of an individual who has not consumed excessive alcohol. Non-alcoholic steatohepatitis (NASH), a severe form of NAFLD, can progress to hepatic cirrhosis and/or hepatocellular carcinoma (HCC). NAFLD is considered to be a hepatic manifestation of metabolic syndrome, and its incidence has risen worldwide in lockstep with the increased global prevalence of obesity. Over the last decade, rodent studies have yielded an impressive list of molecules associated with NAFLD and NASH pathogenesis. However, the identification of currently unknown metabolic factors using mammalian model organisms is inefficient and expensive compared with studies using fish models such as zebrafish (Danio rerio) and medaka (Oryzias latipes). Substantial advances in unraveling the molecular pathogenesis of NAFLD have recently been achieved through unbiased forward genetic screens using small fish models. Furthermore, these easily manipulated organisms have been used to great advantage to evaluate the therapeutic effectiveness of various chemical compounds for the treatment of NAFLD. In this Review, we summarize aspects of NAFLD (specifically focusing on NASH) pathogenesis that have been previously revealed by rodent models, and discuss how small fish are increasingly being used to uncover factors that contribute to normal hepatic lipid metabolism. We describe the various types of fish models in use for this purpose, including those generated by mutation, transgenesis, or dietary or chemical treatment, and contrast them with rodent models. The use of small fish in identifying novel potential therapeutic agents for the treatment of NAFLD and NASH is also addressed.


Asunto(s)
Hígado Graso/patología , Peces/metabolismo , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Peces/genética , Humanos , Enfermedad del Hígado Graso no Alcohólico
16.
J Biol Chem ; 286(51): 43972-43983, 2011 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-21971049

RESUMEN

Autotaxin (ATX) is a multifunctional ecto-type phosphodiesterase that converts lysophospholipids, such as lysophosphatidylcholine, to lysophosphatidic acid (LPA) by its lysophospholipase D activity. LPA is a lipid mediator with diverse biological functions, most of which are mediated by G protein-coupled receptors specific to LPA (LPA1-6). Recent studies on ATX knock-out mice revealed that ATX has an essential role in embryonic blood vessel formation. However, the underlying molecular mechanisms remain to be solved. A data base search revealed that ATX and LPA receptors are conserved in wide range of vertebrates from fishes to mammals. Here we analyzed zebrafish ATX (zATX) and LPA receptors both biochemically and functionally. zATX, like mammalian ATX, showed lysophospholipase D activity to produce LPA. In addition, all zebrafish LPA receptors except for LPA5a and LPA5b were found to respond to LPA. Knockdown of zATX in zebrafish embryos by injecting morpholino antisense oligonucleotides (MOs) specific to zATX caused abnormal blood vessel formation, which has not been observed in other morphant embryos or mutants with vascular defects reported previously. In ATX morphant embryos, the segmental arteries sprouted normally from the dorsal aorta but stalled in midcourse, resulting in aberrant vascular connection around the horizontal myoseptum. Similar vascular defects were not observed in embryos in which each single LPA receptor was attenuated by using MOs. Interestingly, similar vascular defects were observed when both LPA1 and LPA4 functions were attenuated by using MOs and/or a selective LPA receptor antagonist, Ki16425. These results demonstrate that the ATX-LPA-LPAR axis is a critical regulator of embryonic vascular development that is conserved in vertebrates.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Regulación de la Expresión Génica , Hidrolasas Diéster Fosfóricas/química , Receptores del Ácido Lisofosfatídico/metabolismo , Animales , Células HEK293 , Humanos , Hibridación in Situ , Lisofosfolípidos/metabolismo , Ratones , Microscopía Fluorescente/métodos , Neovascularización Fisiológica , Especificidad por Sustrato , Factores de Tiempo , Pez Cebra
17.
J Biochem ; 148(4): 393-401, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20801953

RESUMEN

Mitogen-activated protein kinase kinases (MAPKKs) are important components of the stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) signalling pathway. Two MAPKKs that are crucial transducers upstream of JNK signalling are MKK4 and MKK7. These two MAPKKs directly phosphorylate specific Tyr and Thr residues located in the activation loop of the JNK protein and activate this kinase in response to environmental stress, pro-inflammatory cytokines or developmental cues. Although much is known about the biochemical and structural bases of the catalytic mechanism of the MAPKKs, the regulation and physiological functions of these enzymes during early embryogenesis have remained a mystery until relatively recently. Studies employing a range of animal models have now revealed the essential roles that MAPKKs play in diverse developmental contexts, including in dorsoventral patterning, convergent extension and somitogenesis. Focusing primarily on extensive work done in mouse and zebrafish models, this review summarizes the functional properties of MKK4 and MKK7 during vertebrate and invertebrate development, and the mechanisms by which these kinases regulate multiple steps in the establishment of the body plan of an organism.


Asunto(s)
Desarrollo Embrionario/fisiología , MAP Quinasa Quinasa 4/metabolismo , MAP Quinasa Quinasa 7/metabolismo , Animales , Activación Enzimática , Humanos , Isoenzimas/química , Isoenzimas/clasificación , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa 4/química , MAP Quinasa Quinasa 4/clasificación , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 7/química , MAP Quinasa Quinasa 7/clasificación , MAP Quinasa Quinasa 7/genética , Sistema de Señalización de MAP Quinasas/fisiología , Modelos Moleculares , Filogenia
18.
J Cell Biochem ; 110(4): 1022-37, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20564202

RESUMEN

Stress-induced Sapk/Jnk signaling is involved in cell survival and apoptosis. Recent studies have increased our understanding of the physiological roles of Jnk signaling in embryonic development. However, still unclear is the precise function of Jnk signaling during gastrulation, a critical step in the establishment of the vertebrate body plan. Here we use morpholino-mediated knockdown of the zebrafish orthologs of the Jnk activators Mkk4 and Mkk7 to examine the effect of Jnk signaling abrogation on early vertebrate embryogenesis. Depletion of zebrafish Mkk4b led to abnormal convergent extension (CE) during gastrulation, whereas Mkk7 morphants exhibited defective somitogenesis. Surprisingly, Mkk4b morphants displayed marked upregulation of wnt11, which is the triggering ligand of CE and stimulates Jnk activation via the non-canonical Wnt pathway. Conversely, ectopic activation of Jnk signaling by overexpression of an active form of Mkk4b led to wnt11 downregulation. Mosaic lineage tracing studies revealed that Mkk4b-Jnk signaling suppressed wnt11 expression in a non-cell-autonomous manner. These findings provide the first evidence that wnt11 itself is a downstream target of the Jnk cascade in the non-canonical Wnt pathway. Our work demonstrates that Jnk activation is indispensable for multiple steps during vertebrate body plan formation. Furthermore, non-canonical Wnt signaling may coordinate vertebrate CE movements by triggering Jnk activation that represses the expression of the CE-triggering ligand wnt11.


Asunto(s)
Gástrula , Regulación del Desarrollo de la Expresión Génica , MAP Quinasa Quinasa 4/metabolismo , Transducción de Señal , Proteínas Wnt/genética , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Animales , Secuencia de Bases , Clonación Molecular , Cartilla de ADN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
19.
Biochem Biophys Res Commun ; 396(4): 887-93, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20457130

RESUMEN

Cilia are microtubule-based organelles that are present on the surfaces of almost all vertebrate cells. Most cilia function as sensory or molecular transport structures. Malfunctions of cilia have been implicated in several diseases of human development. The assembly of cilia is initiated by the centriole (or basal body), and several centrosomal proteins are involved in this process. The mammalian LIM protein Ajuba is a well-studied centrosomal protein that regulates cell division but its role in ciliogenesis is unknown. In this study, we isolated the medaka homolog of Ajuba and showed that Ajuba localizes to basal bodies of cilia in growth-arrested cells. Knockdown of Ajuba resulted in randomized left-right organ asymmetries and altered expression of early genes responsible for left-right body axis determination. At the cellular level, we found that Ajuba function was essential for ciliogenesis in the cells lining Kupffer's vesicle; it is these cells that induce the asymmetric fluid flow required for left-right axis determination. Taken together, our findings identify a novel role for Ajuba in the regulation of vertebrate ciliogenesis and left-right axis determination.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Microtúbulos/fisiología , Oryzias/embriología , Animales , Cilios/genética , Cilios/metabolismo , Cilios/fisiología , Clonación Molecular , Técnicas de Silenciamiento del Gen , Corazón/embriología , Proteínas de Homeodominio/genética , Hígado/anomalías , Microtúbulos/genética , Microtúbulos/metabolismo , Oryzias/genética , Oryzias/metabolismo , Bazo/anomalías
20.
Stem Cells Dev ; 19(11): 1723-34, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20412016

RESUMEN

Many studies have shown that it is possible to use culture conditions to direct the differentiation of murine embryonic stem (ES) cells into a variety of cell types, including cardiomyocytes and neurons. However, the molecular mechanisms that control lineage commitment decisions by ES cells remain poorly understood. In this study, we investigated the role of the 3 major mitogen-activated protein kinases (MAPKs: extracellular signal-regulated kinase, c-Jun N-terminal kinase, and p38) in ES cell lineage commitment and showed that the p38 MAPK-specific inhibitor SB203580 blocks the spontaneous differentiation of ES cells into cardiomyocytes and instead induces the differentiation of these ES cells into neurons. Robust p38 MAPK activity between embryoid body culture days 3 and 4 is crucial for cardiomyogenesis of ES cells, and specific inhibition of p38 MAPK activity at this time results in ES cell differentiation into neurons rather than cardiomyocytes. At the molecular level, inhibition of p38 MAPK activity suppresses the expression of bmp-2 mRNA, whereas treatment of ES cells with bone morphogenetic protein 2 (BMP-2) inhibits the neurogenesis induced by SB203580. Further, luciferase reporter assays and chromatin immunoprecipitation experiments showed that BMP-2 expression in ES cells is regulated directly by the transcription factor myocyte enhancer factor 2C, a well-known substrate of p38 MAPK. Our findings reveal the molecular mechanism by which p38 MAPK activity in ES cells drives their commitment to differentiate preferentially into cardiomyocytes, and the conditions under which these same cells might develop into neurons.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Células Madre Embrionarias/fisiología , Miocitos Cardíacos/fisiología , Factores Reguladores Miogénicos/metabolismo , Neuronas/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Proteína Morfogenética Ósea 2/genética , Diferenciación Celular/fisiología , Linaje de la Célula , Células Cultivadas , Células Madre Embrionarias/citología , Inhibidores Enzimáticos/metabolismo , Humanos , Imidazoles/metabolismo , Factores de Transcripción MEF2 , Ratones , Miocitos Cardíacos/citología , Factores Reguladores Miogénicos/genética , Neuronas/citología , Piridinas/metabolismo , Transcripción Genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...