Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Comput Biol ; 20(2): e1011270, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38324613

RESUMEN

CyVerse, the largest publicly-funded open-source research cyberinfrastructure for life sciences, has played a crucial role in advancing data-driven research since the 2010s. As the technology landscape evolved with the emergence of cloud computing platforms, machine learning and artificial intelligence (AI) applications, CyVerse has enabled access by providing interfaces, Software as a Service (SaaS), and cloud-native Infrastructure as Code (IaC) to leverage new technologies. CyVerse services enable researchers to integrate institutional and private computational resources, custom software, perform analyses, and publish data in accordance with open science principles. Over the past 13 years, CyVerse has registered more than 124,000 verified accounts from 160 countries and was used for over 1,600 peer-reviewed publications. Since 2011, 45,000 students and researchers have been trained to use CyVerse. The platform has been replicated and deployed in three countries outside the US, with additional private deployments on commercial clouds for US government agencies and multinational corporations. In this manuscript, we present a strategic blueprint for creating and managing SaaS cyberinfrastructure and IaC as free and open-source software.


Asunto(s)
Inteligencia Artificial , Programas Informáticos , Humanos , Nube Computacional , Edición
2.
Mol Biol Evol ; 40(9)2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37695804

RESUMEN

Uric acid is the main means of nitrogen excretion in uricotelic vertebrates (birds and reptiles) and the end product of purine catabolism in humans and a few other mammals. While uricase is inactivated in mammals unable to degrade urate, the presence of orthologous genes without inactivating mutations in avian and reptilian genomes is unexplained. Here we show that the Gallus gallus gene we name cysteine-rich urate oxidase (CRUOX) encodes a functional protein representing a unique case of cysteine enrichment in the evolution of vertebrate orthologous genes. CRUOX retains the ability to catalyze urate oxidation to hydrogen peroxide and 5-hydroxyisourate (HIU), albeit with a 100-fold reduced efficiency. However, differently from all uricases hitherto characterized, it can also facilitate urate regeneration from HIU, a catalytic property that we propose depends on its enrichment in cysteine residues. X-ray structural analysis highlights differences in the active site compared to known orthologs and suggests a mechanism for cysteine-mediated self-aggregation under H2O2-oxidative conditions. Cysteine enrichment was concurrent with the transition to uricotelism and a shift in gene expression from the liver to the skin where CRUOX is co-expressed with ß-keratins. Therefore, the loss of urate degradation in amniotes has followed opposite evolutionary trajectories: while uricase has been eliminated by pseudogenization in some mammals, it has been repurposed as a redox-sensitive enzyme in the reptilian skin.


Asunto(s)
Cisteína , Reptiles , Piel , Urato Oxidasa , Animales , Cisteína/genética , Peróxido de Hidrógeno , Piel/enzimología , Urato Oxidasa/genética , Urato Oxidasa/metabolismo , Ácido Úrico , Pollos/genética , Reptiles/genética , Reptiles/metabolismo
3.
Nat Ecol Evol ; 4(9): 1239-1246, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32601391

RESUMEN

Among amniotes, reptiles and mammals are differently adapted to terrestrial life. It is generally appreciated that terrestrialization required adaptive changes of vertebrate metabolism, particularly in the mode of nitrogen excretion. However, the current paradigm is that metabolic adaptation to life on land did not involve synthesis of enzymatic pathways de novo, but rather repurposing of existing ones. Here, by comparing the inventory of pyridoxal 5'-phosphate-dependent enzymes in different amniotes, we identify in silico a pathway for sulfur metabolism present in chick embryos but not in mammals. Cysteine lyase contains haem and pyridoxal 5'-phosphate co-factors and converts cysteine and sulfite into cysteic acid and hydrogen sulfide, respectively. A specific cysteic acid decarboxylase produces taurine, while hydrogen sulfide is recycled into cysteine by cystathionine beta-synthase. This reaction sequence enables the formation of sulfonated amino acids during embryo development in the egg at no cost of reduced sulfur. The pathway originated around 300 million years ago in a proto-reptile by cystathionine beta-synthase duplication, cysteine lyase neofunctionalization and cysteic acid decarboxylase co-option. Our findings indicate that adaptation to terrestrial life involved innovations in metabolic pathways, and reveal the molecular mechanisms by which such innovations arose in amniote evolution.


Asunto(s)
Cistationina gamma-Liasa , Sulfuro de Hidrógeno , Animales , Embrión de Pollo , Cistationina betasintasa/genética , Cisteína , Azufre
4.
J Hered ; 110(2): 194-210, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30597046

RESUMEN

Next-generation sequencing (NGS) and expression technologies were utilized to investigate the genes and sequence elements in a 586 kb region of chicken chromosome 1 associated with the autosomal recessive diplopodia-1 (dp-1) mutation. This mutation shows a syndromic phenotype similar to known human developmental abnormalities (e.g., cleft palate, polydactyly, omphalocele [exposed viscera]). Toward our goal to ascertain the variant responsible, the entire 586 kb region was sequenced following utilization of a specifically designed capture array and to confirm/validate fine-mapping results. Bioinformatic analyses identified a total of 6142 sequence variants, which included SNPs, indels, and gaps. Of these, 778 SNPs, 146 micro-indels, and 581 gaps were unique to the UCD-Dp-1.003 inbred congenic line; those found within exons and splice sites were studied for contribution to the mutant phenotype. Upon further validation with additional mutant samples, a smaller subset (of variants [51]) remains linked to the mutation. Additionally, utilization of specific samples in the NGS technology was advantageous in that fine-mapping methodologies eliminated an additional 326 kb of sequence information on chromosome 1. Predicted and confirmed protein-coding genes within the smaller 260 kb region were assessed for their developmental expression patterns over several stages of early embryogenesis in regions/tissues of interest (e.g., digits, craniofacial region). Based on these results and known function in other vertebrates, 2 genes within 5 kb of each other, MRE11 and GPR83, are proposed as high-priority candidates for the dp-1 mutation.


Asunto(s)
Pollos/genética , Anomalías Craneofaciales/genética , Deformidades Congénitas de las Extremidades/genética , Proteína Homóloga de MRE11/genética , Receptores Acoplados a Proteínas G/genética , Animales , Mapeo Cromosómico , Anomalías Craneofaciales/diagnóstico , Análisis Mutacional de ADN , Secuenciación de Nucleótidos de Alto Rendimiento , Deformidades Congénitas de las Extremidades/diagnóstico , Mutación , Síndrome
5.
Dev Dyn ; 246(12): 969, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29144604
8.
PLoS One ; 11(4): e0153591, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27097030

RESUMEN

Epithelial-mesenchymal transition (EMT) is an evolutionarily conserved process during which cells lose epithelial characteristics and gain a migratory phenotype. Although downregulation of epithelial cadherins by Snail and other transcriptional repressors is generally considered a prerequisite for EMT, recent studies have challenged this view. Here we investigate the relationship between E-cadherin and P-cadherin expression and localization, Snail function and EMT during gastrulation in chicken embryos. Expression analyses show that while E-cadherin transcripts are detected in the epiblast but not in the primitive streak or mesoderm, P-cadherin mRNA and protein are present in the epiblast, primitive and mesoderm. Antibodies that specifically recognize E-cadherin are not presently available. During EMT, P-cadherin relocalizes from the lateral surfaces of epithelial epiblast cells to a circumferential distribution in emerging mesodermal cells. Cells electroporated with an E-cadherin expression construct undergo EMT and migrate into the mesoderm. An examination of Snail function showed that reduction of Slug (SNAI2) protein levels using a morpholino fails to inhibit EMT, and expression of human or chicken Snail in epiblast cells fails to induce EMT. In contrast, cells expressing the Rho inhibitor peptide C3 rapidly exit the epiblast without activating Slug or the mesoderm marker N-cadherin. Together, these experiments show that epiblast cells undergo EMT while retaining P-cadherin, and raise questions about the mechanisms of EMT regulation during avian gastrulation.


Asunto(s)
Cadherinas/metabolismo , Regulación hacia Abajo , Transición Epitelial-Mesenquimal , Gastrulación , Secuencia de Aminoácidos , Animales , Cadherinas/química , Movimiento Celular , Pollos , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Humanos , Mesodermo/citología , Ratones , Datos de Secuencia Molecular , Transporte de Proteínas , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo
10.
Methods Mol Biol ; 1211: 69-76, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25218377

RESUMEN

In situ hybridization (ISH) in embryos allows the visualization of specific RNAs as a readout of gene expression during normal development or after experimental manipulations. ISH using short DNA probes containing locked nucleic acid nucleotides (LNAs) holds the additional advantage of allowing the detection of specific RNA splice variants or of closely related family members that differ in only short regions, creating new diagnostic and detection opportunities. Here we describe methods for using short (14-24 nt) DNA probes containing LNA nucleotides to detect moderately to highly expressed RNAs in whole chick embryos during the first 5 days of embryonic development. The protocol is easily adaptable for use with embryos of other vertebrate species.


Asunto(s)
Embrión de Pollo/metabolismo , Sondas de ADN/análisis , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ/métodos , Oligonucleótidos/análisis , ARN Mensajero/análisis , Animales , Sondas de ADN/genética , Oligonucleótidos/genética , ARN Mensajero/genética
11.
Nucleic Acids Res ; 42(Database issue): D933-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24150938

RESUMEN

GEISHA (Gallus Expression In Situ Hybridization Analysis; http://geisha.arizona.edu) is an in situ hybridization gene expression and genomic resource for the chicken embryo. This update describes modifications that enhance its utility to users. During the past 5 years, GEISHA has undertaken a significant restructuring to more closely conform to the data organization and formatting of Model Organism Databases in other species. This has involved migrating from an entry-centric format to one that is gene-centered. Database restructuring has enabled the inclusion of data pertaining to chicken genes and proteins and their orthologs in other species. This new information is presented through an updated user interface. In situ hybridization data in mouse, frog, zebrafish and fruitfly are integrated with chicken genomic and expression information. A resource has also been developed that integrates the GEISHA interface information with the Online Mendelian Inheritance in Man human disease gene database. Finally, the Chicken Gene Nomenclature Committee database and the GEISHA database have been integrated so that they draw from the same data resources.


Asunto(s)
Embrión de Pollo/metabolismo , Pollos/genética , Bases de Datos Genéticas , Expresión Génica , Animales , Genómica , Hibridación in Situ , Internet , Ratones , Modelos Animales , ARN Mensajero/análisis
12.
Dev Dyn ; 243(3): 497-508, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24166734

RESUMEN

BACKGROUND: Transforming growth factor-beta (TGFß) signaling regulates a myriad of biological processes during embryogenesis, in the adult, and during the manifestation of disease. TGFß signaling is propagated through one of three TGFß ligands interacting with Type I and Type II receptors, and Type III co-receptors. Although TGFß signaling is regulated partly by the combinatorial expression patterns of TGFß receptors and ligands, a comprehensive gene expression analysis has not been published. RESULTS: Here we report the embryonic mRNA expression patterns in chicken embryos of the canonical TGFß ligands (TGFB1, TGFB2, and TGFB3) and receptors (TGFBR1, TGFBR2, TGFBR3), plus the Activin A receptor, type 1 (ACVR1) and co receptor Endoglin (ENG) that also transduce TGFß signaling. CONCLUSIONS: TGFB ligands and receptors show dynamic and frequently overlapping expression patterns in numerous embryonic cell layers and structures. Integrating expression information identifies combinations of ligands and receptors that are involved in specific developmental processes including somitogenesis, cardiogenesis and vasculogenesis.


Asunto(s)
Proteínas Aviares/biosíntesis , Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Factor de Crecimiento Transformador beta/biosíntesis , Animales , Antígenos CD/biosíntesis , Antígenos CD/genética , Proteínas Aviares/genética , Embrión de Pollo , Receptores de Factores de Crecimiento Transformadores beta/genética , Factor de Crecimiento Transformador beta/genética
14.
Dis Model Mech ; 6(3): 632-42, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23264563

RESUMEN

Endothelia in the atrioventricular (AV) canal of the developing heart undergo a prototypical epithelial mesenchymal transition (EMT) to begin heart valve formation. Using an in vitro invasion assay, an extracellular matrix protein, Olfactomedin-1 (OLFM1), was found to increase mesenchymal cell numbers in AV canals from embryonic chick hearts. Treatment with both anti-OLFM1 antibody and siRNA targeting OLFM1 inhibits mesenchymal cell formation. OLFM1 does not alter cell proliferation, migration or apoptosis. Dispersion, but lack of invasion in the presence of inhibiting antibody, identifies a specific role for OLFM1 in cell invasion during EMT. This role is conserved in other epithelia, as OLFM1 similarly enhances invasion by MDCK epithelial cells in a transwell assay. Synergy is observed when TGFß2 and OLFM1 are added to MDCK cell cultures, indicating that OLFM-1 activity is cooperative with TGFß. Inhibition of both OLFM1 and TGFß in heart invasion assays shows a similar cooperative role during development. To explore OLFM1 activity during EMT, representative EMT markers were examined. Effects of OLFM1 protein and anti-OLFM1 on transcripts of cell-cell adhesion molecules and the transcription factors Snail-1, Snail-2, Twist1 and Sox-9 argue that OLFM1 does not initiate EMT. Rather, regulation of transcripts of Zeb1 and Zeb2, secreted proteases and mesenchymal cell markers by both OLFM1 and anti-OLFM1 is consistent with regulation of the cell invasion step of EMT. We conclude that OLFM1 is present and necessary during EMT in the embryonic chick heart. Its role in cell invasion and mesenchymal cell gene regulation suggests an invasion checkpoint in EMT where OLFM1 acts to promote cell invasion into the three-dimensional matrix.


Asunto(s)
Movimiento Celular , Transición Epitelial-Mesenquimal , Proteínas de la Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Corazón/embriología , Animales , Anticuerpos/farmacología , Biomarcadores/metabolismo , Muerte Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Perros , Transición Epitelial-Mesenquimal/efectos de los fármacos , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/inmunología , Glicoproteínas/genética , Glicoproteínas/inmunología , Corazón/efectos de los fármacos , Humanos , Células de Riñón Canino Madin Darby , Mesodermo/citología , Mesodermo/efectos de los fármacos , Mesodermo/embriología , Miocardio/citología , Miocardio/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Factor de Crecimiento Transformador beta/farmacología
15.
J Biol Chem ; 287(46): 38505-14, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22995917

RESUMEN

FGF signaling plays a pivotal role in regulating cell movements and lineage induction during gastrulation. Here we identify 44 microRNAs that are expressed in the primitive streak region of gastrula stage chicken embryos. We show that the primary effect of FGF signaling on microRNA abundance is to negatively regulate the levels of miR-let-7b, -9, -19b, -107, -130b, and -218. LIN28B inhibits microRNA processing and is positively regulated by FGF signaling. Gain- and loss-of-function experiments show that LIN28B negatively regulates the expression of miR-19b, -130b, and let-7b, whereas negative modulation of miR-9, -107, and -218 appears to be independent of LIN28B function. Predicted mRNA targets of the FGF-regulated microRNAs are over-represented in serine/threonine and tyrosine kinase receptors, including ACVR1, ACVR2B, PDGFRA, TGFBR1, and TGFBR3. Luciferase assays show that these and other candidates are targeted by FGF-regulated microRNAs. PDGFRA, a receptor whose activity is required for cell migration through the primitive streak, is a target of miR-130b and -218 in vivo. These results identify a novel mechanism by which FGF signaling regulates gene expression by negatively modulating microRNA abundance through both LIN28B-dependent and LIN28B-independent pathways.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Represoras/metabolismo , Animales , Tipificación del Cuerpo/genética , Movimiento Celular , Embrión de Pollo , Proteínas de Unión al ADN/metabolismo , Gástrula/metabolismo , Gastrulación , Regulación del Desarrollo de la Expresión Génica , Células HeLa , Humanos , Hibridación in Situ , MicroARNs/metabolismo , Conformación de Ácido Nucleico , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal
16.
PLoS One ; 7(7): e41574, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22844503

RESUMEN

BACKGROUND: Recently, MicroRNAs (miR) and AMP-kinase (AMPK) have emerged as prominent players in the development of cardiac hypertrophy and heart failure. We hypothesized that components of the adenosine monophosphate-activated kinase (AMPK) pathway are targeted by miRs and alter AMPK signaling during pathological cardiac stress. METHODOLOGY/PRINCIPAL FINDINGS: Using a mouse model of hypertrophic cardiomyopathy (HCM), we demonstrated early elevation of miR-195 and miR-451 in HCM hearts, which targets MO25, a central component of the MO25/STRAD/LKB1 complex that acts as an upstream kinase for AMPK. We show functional targeting of MO25 by miR-195 and -451. Further in vitro interrogation of MO25 as a functional target validated this hypothesis where over-expression of miR-195 in C2C12 cells knocked down MO25 expression levels and downstream AMPK signaling (phosphorylation of Acetyl CoA carboxylase [ACC] and AMPK activity assay), similar to MO25 knockdown in C2C12 cells by siRNA. Parallel changes were measured in 60 day R403Q HCM male hearts that were rescued by short-term administration of AICAR, an AMPK agonist. CONCLUSIONS/SIGNIFICANCE: Elevated miR-195 targets the LKB1/AMPK signaling axis in HCM progression and implicates a functional role in HCM disease progression. MiR-195 may serve as potential therapeutics or therapeutic targets for heart disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenilato Quinasa/metabolismo , MicroARNs/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/genética , Proteínas Quinasas Activadas por AMP , Animales , Secuencia de Bases , Proteínas de Unión al Calcio , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/patología , Línea Celular , Progresión de la Enfermedad , Activación Enzimática/genética , Humanos , Masculino , Ratones , MicroARNs/genética , Terapia Molecular Dirigida , Miocardio/metabolismo , Miocardio/patología , Especificidad de Órganos , Regulación hacia Arriba/genética
17.
Proc Natl Acad Sci U S A ; 108(48): 19240-5, 2011 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-22084120

RESUMEN

Neural crest stem cells can be isolated from differentiated cultures of human pluripotent stem cells, but the process is inefficient and requires cell sorting to obtain a highly enriched population. No specific method for directed differentiation of human pluripotent cells toward neural crest stem cells has yet been reported. This severely restricts the utility of these cells as a model for disease and development and for more applied purposes such as cell therapy and tissue engineering. In this report, we use small-molecule compounds in a single-step method for the efficient generation of self-renewing neural crest-like stem cells in chemically defined media. This approach is accomplished directly from human pluripotent cells without the need for coculture on feeder layers or cell sorting to obtain a highly enriched population. Critical to this approach is the activation of canonical Wnt signaling and concurrent suppression of the Activin A/Nodal pathway. Over 12-14 d, pluripotent cells are efficiently specified along the neuroectoderm lineage toward p75(+) Hnk1(+) Ap2(+) neural crest-like cells with little or no contamination by Pax6(+) neural progenitors. This cell population can be clonally amplified and maintained for >25 passages (>100 d) while retaining the capacity to differentiate into peripheral neurons, smooth muscle cells, and mesenchymal precursor cells. Neural crest-like stem cell-derived mesenchymal precursors have the capacity for differentiation into osteocytes, chondrocytes, and adipocytes. In sum, we have developed methods for the efficient generation of self-renewing neural crest stem cells that greatly enhance their potential utility in disease modeling and regenerative medicine.


Asunto(s)
Diferenciación Celular/fisiología , Cresta Neural/citología , Células Madre Pluripotentes/citología , Transducción de Señal/fisiología , Proteínas Smad/metabolismo , Ingeniería de Tejidos/métodos , Proteínas Wnt/metabolismo , Western Blotting , Técnicas de Cultivo de Célula , Humanos , Microscopía Fluorescente , Reacción en Cadena en Tiempo Real de la Polimerasa
18.
BMC Evol Biol ; 11: 302, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21999483

RESUMEN

BACKGROUND: The forkhead transcription factor gene E1 (FOXE1) plays an important role in regulation of thyroid development, palate formation and hair morphogenesis in mammals. However, avian FOXE1 genes have not been characterized and as such, codon evolution of FOXE1 orthologs in a broader evolutionary context of mammals and birds is not known. RESULTS: In this study we identified the avian FOXE1 gene in chicken, turkey and zebra finch, all of which consist of a single exon. Chicken and zebra finch FOXE1 are uniquely located on the sex-determining Z chromosome. In situ hybridization shows that chicken FOXE1 is specifically expressed in the developing thyroid. Its expression is initiated at the placode stage and is maintained during the stages of vesicle formation and follicle primordia. Based on this expression pattern, we propose that avian FOXE1 may be involved in regulating the evagination and morphogenesis of thyroid. Chicken FOXE1 is also expressed in growing feathers. Sequence analysis identified two microdeletions in the avian FOXE1 genes, corresponding to the loss of a transferable repression domain and an engrailed homology motif 1 (Eh1) C-terminal to the forkhead domain. The avian FOXE1 proteins exhibit a significant sequence divergence of the C-terminus compared to those of amphibian and mammalian FOXE1. The codon evolution analysis (dN/dS) of FOXE1 shows a significantly increased dN/dS ratio in the avian lineages, consistent with either a relaxed purifying selection or positive selection on a few residues in avian FOXE1 evolution. Further site specific analysis indicates that while relaxed purifying selection is likely to be a predominant cause of accelerated evolution at the 3'-region of avian FOXE1, a few residues might have evolved under positive selection. CONCLUSIONS: We have identified three avian FOXE1 genes based on synteny and sequence similarity as well as characterized the expression pattern of the chicken FOXE1 gene during development. Our evolutionary analyses suggest that while a relaxed purifying selection is likely to be the dominant force driving accelerated evolution of avian FOXE1 genes, a few residues may have evolved adaptively. This study provides a basis for future genetic and comparative biochemical studies of FOXE1.


Asunto(s)
Pollos/genética , Evolución Molecular , Pinzones/genética , Factores de Transcripción Forkhead/genética , Selección Genética , Pavos/genética , Región de Flanqueo 3'/genética , Animales , Secuencia de Bases , Cartilla de ADN/genética , Plumas/crecimiento & desarrollo , Plumas/metabolismo , Factores de Transcripción Forkhead/metabolismo , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Eliminación de Secuencia , Glándula Tiroides/crecimiento & desarrollo , Glándula Tiroides/metabolismo
19.
BMC Dev Biol ; 11: 20, 2011 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-21418646

RESUMEN

BACKGROUND: FGF signalling regulates numerous aspects of early embryo development. During gastrulation in amniotes, epiblast cells undergo an epithelial to mesenchymal transition (EMT) in the primitive streak to form the mesoderm and endoderm. In mice lacking FGFR1, epiblast cells in the primitive streak fail to downregulate E-cadherin and undergo EMT, and cell migration is inhibited. This study investigated how FGF signalling regulates cell movement and gene expression in the primitive streak of chicken embryos. RESULTS: We find that pharmacological inhibition of FGFR activity blocks migration of cells through the primitive streak of chicken embryos without apparent alterations in the level or intracellular localization of E-cadherin. E-cadherin protein is localized to the periphery of epiblast, primitive streak and some mesodermal cells. FGFR inhibition leads to downregulation of a large number of regulatory genes in the preingression epiblast adjacent to the primitive streak, the primitive streak and the newly formed mesoderm. This includes members of the FGF, NOTCH, EPH, PDGF, and canonical and non-canonical WNT pathways, negative modulators of these pathways, and a large number of transcriptional regulatory genes. SNAI2 expression in the primitive streak and mesoderm is not altered by FGFR inhibition, but is downregulated only in the preingression epiblast region with no significant effect on E-cadherin. Furthermore, over expression of SNAIL has no discernable effect on E-cadherin protein levels or localization in epiblast, primitive streak or mesodermal cells. FGFR activity modulates distinct downstream pathways including RAS/MAPK and PI3K/AKT. Pharmacological inhibition of MEK or AKT indicate that these downstream effectors control discrete and overlapping groups of genes during gastrulation. FGFR activity regulates components of several pathways known to be required for cell migration through the streak or in the mesoderm, including RHOA, the non-canonical WNT pathway, PDGF signalling and the cell adhesion protein N-cadherin. CONCLUSIONS: In chicken embryos, FGF signalling regulates cell movement through the primitive streak by mechanisms that appear to be independent of changes in E-cadherin expression or protein localization. The positive and negative effects on large groups of genes by pharmacological inhibition of FGF signalling, including major signalling pathways and transcription factor families, indicates that the FGF pathway is a focal point of regulation during gastrulation in chicken.


Asunto(s)
Cadherinas/genética , Movimiento Celular , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Línea Primitiva/metabolismo , Proteínas ras/metabolismo , Animales , Western Blotting , Cadherinas/metabolismo , Embrión de Pollo , Electroporación , Factores de Crecimiento de Fibroblastos/genética , Gastrulación , Expresión Génica , Hibridación in Situ , Análisis por Micromatrices , Proteínas Quinasas Activadas por Mitógenos/genética , Fosfatidilinositol 3-Quinasas/genética , Reacción en Cadena de la Polimerasa , Línea Primitiva/embriología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas ras/genética
20.
J Cell Sci ; 123(Pt 18): 3136-45, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20736303

RESUMEN

Regulation of actin filament assembly is essential for efficient contractile activity in striated muscle. Leiomodin is an actin-binding protein and homolog of the pointed-end capping protein, tropomodulin. These proteins are structurally similar, sharing a common domain organization that includes two actin-binding sites. Leiomodin also contains a unique C-terminal extension that has a third actin-binding WH2 domain. Recently, the striated-muscle-specific isoform of leiomodin (Lmod2) was reported to be an actin nucleator in cardiomyocytes. Here, we have identified a function of Lmod2 in the regulation of thin filament lengths. We show that Lmod2 localizes to the pointed ends of thin filaments, where it competes for binding with tropomodulin-1 (Tmod1). Overexpression of Lmod2 results in loss of Tmod1 assembly and elongation of the thin filaments from their pointed ends. The Lmod2 WH2 domain is required for lengthening because its removal results in a molecule that caps the pointed ends similarly to Tmod1. Furthermore, Lmod2 transcripts are first detected in the heart after it has begun to beat, suggesting that the primary function of Lmod2 is to maintain thin filament lengths in the mature heart. Thus, Lmod2 antagonizes the function of Tmod1, and together, these molecules might fine-tune thin filament lengths.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas de Microfilamentos/metabolismo , Miocardio/metabolismo , Tropomiosina/antagonistas & inhibidores , Tropomiosina/metabolismo , Citoesqueleto de Actina/química , Animales , Células Cultivadas , Embrión de Pollo , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Células Musculares/metabolismo , Miocardio/química , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Tropomiosina/química , Tropomiosina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...