Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Elife ; 122023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38108810

RESUMEN

The enteric nervous system (ENS), a collection of neural cells contained in the wall of the gut, is of fundamental importance to gastrointestinal and systemic health. According to the prevailing paradigm, the ENS arises from progenitor cells migrating from the neural crest and remains largely unchanged thereafter. Here, we show that the lineage composition of maturing ENS changes with time, with a decline in the canonical lineage of neural-crest derived neurons and their replacement by a newly identified lineage of mesoderm-derived neurons. Single cell transcriptomics and immunochemical approaches establish a distinct expression profile of mesoderm-derived neurons. The dynamic balance between the proportions of neurons from these two different lineages in the post-natal gut is dependent on the availability of their respective trophic signals, GDNF-RET and HGF-MET. With increasing age, the mesoderm-derived neurons become the dominant form of neurons in the ENS, a change associated with significant functional effects on intestinal motility which can be reversed by GDNF supplementation. Transcriptomic analyses of human gut tissues show reduced GDNF-RET signaling in patients with intestinal dysmotility which is associated with reduction in neural crest-derived neuronal markers and concomitant increase in transcriptional patterns specific to mesoderm-derived neurons. Normal intestinal function in the adult gastrointestinal tract therefore appears to require an optimal balance between these two distinct lineages within the ENS.


Asunto(s)
Sistema Nervioso Entérico , Factor Neurotrófico Derivado de la Línea Celular Glial , Adulto , Humanos , Motilidad Gastrointestinal , Perfilación de la Expresión Génica , Mesodermo
2.
Proc Natl Acad Sci U S A ; 120(4): e2217687120, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36649430

RESUMEN

The heart develops in a synchronized sequence of proliferation and differentiation of cardiac progenitor cells (CPCs) from two anatomically distinct pools of cells, the first heart field (FHF) and second heart field (SHF). Congenital heart defects arise upon dysregulation of these processes, many of which are restricted to derivatives of the FHF or SHF. Of the conserved set of signaling pathways that regulate development, the Wnt signaling pathway has long been known for its importance in SHF development. The source of such Wnts has remained elusive, though it has been postulated that these Wnts are secreted from ectodermal or endodermal sources. The central question remains unanswered: Where do these Wnts come from? Here, we show that CPCs autoregulate SHF development via Wnt through genetic manipulation of a key Wnt export protein (Wls), scRNA-seq analysis of CPCs, and use of our precardiac organoid system. Through this, we identify dysregulated developmental trajectories of anterior SHF cell fate, leading to a striking single ventricle phenotype in knockout embryos. We then applied our findings to our precardiac organoid model and found that Wnt2 is sufficient to restore SHF cell fate in our model of disrupted endogenous Wnt signaling. In this study, we provide a basis for SHF cell fate decision-proliferation vs. differentiation-autoregulated by CPCs through Wnt.


Asunto(s)
Cardiopatías Congénitas , Corazón , Humanos , Corazón/fisiología , Diferenciación Celular , Vía de Señalización Wnt , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Regulación del Desarrollo de la Expresión Génica
3.
Differentiation ; 130: 7-15, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36527791

RESUMEN

Fibroblast growth factors (Fgfs) have long been implicated in processes critical to embryonic development, such as cell survival, migration, and differentiation. Several mouse models of organ development ascribe a prosurvival requirement specifically to FGF8. Here, we explore the potential role of prosurvival FGF8 signaling in kidney development. We have previously demonstrated that conditional deletion of Fgf8 in the mesodermal progenitors that give rise to the kidney leads to renal aplasia in the mutant neonate. Deleterious consequences caused by loss of FGF8 begin to manifest by E14.5 when massive aberrant cell death occurs in the cortical nephrogenic zone in the rudimentary kidney as well as in the renal vesicles that give rise to the nephrons. To rescue cell death in the Fgf8 mutant kidney, we inactivate the genes encoding the pro-apoptotic factors BAK and BAX. In a wild-type background, the loss of Bak and Bax abrogates normal cell death and has minimal effect on renal development. However, in Fgf8 mutants, the combined loss of Bak and Bax rescues aberrant cell death in the kidneys and restores some measure of kidney development: 1) the nephron progenitor population is greatly increased; 2) some glomeruli form, which are rarely observed in Fgf8 mutants; and 3) kidney size is rescued by about 50% at E18.5. The development of functional nephrons, however, is not rescued. Thus, FGF8 signaling is required for nephron progenitor survival by regulating BAK/BAX and for subsequent steps involving, as yet, undefined roles in kidney development.


Asunto(s)
Riñón , Nefronas , Ratones , Animales , Femenino , Embarazo , Proteína X Asociada a bcl-2/metabolismo , Nefronas/metabolismo , Apoptosis , Diferenciación Celular , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Factor 8 de Crecimiento de Fibroblastos/metabolismo
4.
Differentiation ; 130: 28-31, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36543010

RESUMEN

The International Society of Differentiation was born from the First International Conference on Cell Differentiation conceived by D.V. and held in Nice, France in 1971. The conference also resulted in the creation of the journal of the Society named Differentiation. The Society advocates for the field of differentiation through the journal Differentiation, organizing and supporting international scientific conferences, honoring scientific achievements, and supporting trainees.


Asunto(s)
Diferenciación Celular , Sociedades Científicas , Sociedades Científicas/historia
5.
Dev Dyn ; 251(5): 877-884, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34719815

RESUMEN

BACKGROUND: Fibroblast growth factors (Fgfs) are required for survival and organ formation during embryogenesis. Fgfs often execute their functions redundantly. Previous analysis of Fgf3 mutants revealed effects on inner ear formation and embryonic survival with incomplete penetrance. RESULTS: Here, we show that presence of a neomycin resistance gene (neo) replacing the Fgf3 coding region leads to reduced survival during embryogenesis and an increased penetrance of inner ear defects. Fgf3neo/neo mutants showed reduced expression of Fgf4, which is positioned in close proximity to the Fgf3 locus in the mouse genome. Conditional inactivation of Fgf4 during inner ear development on a Fgf3 null background using Fgf3/4 cis mice revealed a redundant requirement between these Fgfs during otic placode induction. In contrast, inactivation of Fgf3 and Fgf4 in the pharyngeal region where both Fgfs are also co-expressed using a Foxg1-Cre driver did not affect development of the pharyngeal arches. However, these mutants showed reduced perinatal survival. CONCLUSIONS: These results highlight the importance of Fgf signaling during development. In particular, different members of the Fgf family act redundantly to guarantee inner ear formation and embryonic survival.


Asunto(s)
Oído Interno , Factores de Crecimiento de Fibroblastos , Animales , Ectodermo/metabolismo , Femenino , Factor 3 de Crecimiento de Fibroblastos/genética , Factor 3 de Crecimiento de Fibroblastos/metabolismo , Factor 4 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Transcripción Forkhead/genética , Ratones , Familia de Multigenes , Proteínas del Tejido Nervioso/genética , Embarazo
6.
Elife ; 92020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33210601

RESUMEN

During vertebrate development, the presomitic mesoderm (PSM) periodically segments into somites, which will form the segmented vertebral column and associated muscle, connective tissue, and dermis. The periodicity of somitogenesis is regulated by a segmentation clock of oscillating Notch activity. Here, we examined mouse mutants lacking only Fgf4 or Fgf8, which we previously demonstrated act redundantly to prevent PSM differentiation. Fgf8 is not required for somitogenesis, but Fgf4 mutants display a range of vertebral defects. We analyzed Fgf4 mutants by quantifying mRNAs fluorescently labeled by hybridization chain reaction within Imaris-based volumetric tissue subsets. These data indicate that FGF4 maintains Hes7 levels and normal oscillatory patterns. To support our hypothesis that FGF4 regulates somitogenesis through Hes7, we demonstrate genetic synergy between Hes7 and Fgf4, but not with Fgf8. Our data indicate that Fgf4 is potentially important in a spectrum of human Segmentation Defects of the Vertebrae caused by defective Notch oscillations.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factor 4 de Crecimiento de Fibroblastos/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Embrión de Mamíferos , Factor 4 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Columna Vertebral/anomalías
7.
Biomed Pharmacother ; 132: 110856, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33125970

RESUMEN

PURPOSE: The incidence of retinopathy of prematurity (ROP) has increased continuously in recent years. However, the therapeutic effects of current treatments still remain undesired. This study aims to investigate the role of C-CBL in retinal angiogenesis in ROP and its potential as a therapeutic target. METHODS: Mouse retina microvascular endothelial cells (mRMECs) and induced experimental ROP/ oxygen-induced retinopathy (OIR) mice were employed to investigate the role of C-CBL in angiogenesis with combined molecular and cellular approaches, and histopathology methods. OIR mouse pups at postnatal day 12 (P12) were either injected intravitreally with adenovirus overexpressing c-Cbl or c-Cbl siRNA. Retinal neovascularization and avascular status were evaluated by retinal immunofluorescence (IF) staining, whole-mounts and hematoxylin and eosin (H&E) staining. RESULTS: C-CBL inhibits neovascularization by negatively regulating JAK2/STAT3/VEGF signaling axis in a ubiquitination-dependent manner. Knockdown of c-Cbl by siRNA reduced ubiquitin-mediated JAK2 degradation and increased levels of p-JAK2, p-STAT3, VEGF, and neovascularization in mRMECs, which can be reversed by JAK2 inhibitor treatment. While knockdown of c-Cbl significantly increased neovascular (NV) zone in the retinas, c-Cbl overexpression inhibited neovascularization in the retinal tissues in OIR mice. CONCLUSION: We found that C-CBL is required for anti-neovascularization process in ROP development by inhibiting JAK2/STAT3-dependent angiogenesis. Thus, our finding strongly suggest that C-CBL may be a potential novel therapeutic target for treating ROP.


Asunto(s)
Proteínas Proto-Oncogénicas c-cbl/genética , Neovascularización Retiniana/patología , Retinopatía de la Prematuridad/patología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/patología , Técnicas de Silenciamiento del Gen , Janus Quinasa 2/metabolismo , Ratones , Ratones Endogámicos C57BL , Oxígeno , Neovascularización Retiniana/genética , Vasos Retinianos/citología , Vasos Retinianos/patología , Retinopatía de la Prematuridad/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Development ; 147(21)2020 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-32907848

RESUMEN

The closure of the embryonic ventral body wall in amniotes is an important morphogenetic event and is essential for life. Defects in human ventral wall closure are a major class of birth defect and a significant health burden. Despite this, very little is understood about how the ventral body wall is formed. Here, we show that fibroblast growth factor (FGF) ligands FGF8, FGF17 and FGF18 are essential for this process. Conditional mouse mutants for these genes display subtle migratory defects in the abdominal muscles of the ventral body wall and an enlarged umbilical ring, through which the internal organs are extruded. By refining where and when these genes are required using different Cre lines, we show that Fgf8 and Fgf17 are required in the presomitic mesoderm, whereas Fgf18 is required in the somites. This study identifies complex and multifactorial origins of ventral wall defects and has important implications for understanding their origins during embryonic development.


Asunto(s)
Tipificación del Cuerpo , Embrión de Mamíferos/metabolismo , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Tipificación del Cuerpo/genética , Femenino , Factor 8 de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/genética , Regulación del Desarrollo de la Expresión Génica , Hernia Umbilical , Masculino , Ratones , Modelos Biológicos , Morfogénesis , Mutación/genética , Especificidad de Órganos , Dominios Proteicos , Somitos/metabolismo , Células Madre/metabolismo
9.
Sci Adv ; 6(23): eaaz0742, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32537491

RESUMEN

The positional information theory proposes that a coordinate system provides information to embryonic cells about their position and orientation along a patterning axis. Cells interpret this information to produce the appropriate pattern. During development, morphogens and interpreter transcription factors provide this information. We report a gradient of Meis homeodomain transcription factors along the mouse limb bud proximo-distal (PD) axis antiparallel to and shaped by the inhibitory action of distal fibroblast growth factor (FGF). Elimination of Meis results in premature limb distalization and HoxA expression, proximalization of PD segmental borders, and phocomelia. Our results show that Meis transcription factors interpret FGF signaling to convey positional information along the limb bud PD axis. These findings establish a new model for the generation of PD identities in the vertebrate limb and provide a molecular basis for the interpretation of FGF signal gradients during axial patterning.

10.
J Dev Biol ; 8(2)2020 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-32244588

RESUMEN

The molecular mechanisms regulating neurogenesis involve the control of gene expression by transcription factors. Gbx1 and Gbx2, two members of the Gbx family of homeodomain-containing transcription factors, are known for their essential roles in central nervous system development. The expression domains of mouse Gbx1 and Gbx2 include regions of the forebrain, anterior hindbrain, and spinal cord. In the spinal cord, Gbx1 and Gbx2 are expressed in PAX2+ interneurons of the dorsal horn and ventral motor neuron progenitors. Based on their shared domains of expression and instances of overlap, we investigated the functional relationship between Gbx family members in the developing spinal cord using Gbx1-/-, Gbx2-/-, and Gbx1-/-/Gbx2-/- embryos. In situ hybridization analyses of embryonic spinal cords show upregulation of Gbx2 expression in Gbx1-/- embryos and upregulation of Gbx1 expression in Gbx2-/- embryos. Additionally, our data demonstrate that Gbx genes regulate development of a subset of PAX2+ dorsal inhibitory interneurons. While we observe no difference in overall proliferative status of the developing ependymal layer, expansion of proliferative cells into the anatomically defined mantle zone occurs in Gbx mutants. Lastly, our data shows a marked increase in apoptotic cell death in the ventral spinal cord of Gbx mutants during mid-embryonic stages. While our studies reveal that both members of the Gbx gene family are involved in development of subsets of PAX2+ dorsal interneurons and survival of ventral motor neurons, Gbx1 and Gbx2 are not sufficient to genetically compensate for the loss of one another. Thus, our studies provide novel insight to the relationship harbored between Gbx1 and Gbx2 in spinal cord development.

11.
Dev Dyn ; 248(9): 882-893, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31290205

RESUMEN

BACKGROUND: Fibroblast growth factor 18 (FGF18) functions in the development of several tissues, including the lung, limb bud, palate, skeleton, central nervous system, and hair follicle. Mice containing a germline knockout of Fgf18 (Fgf18 -/- ) die shortly after birth. Postnatally, FGF18 is being evaluated for pathogenic roles in fibrosis and several types of cancer. The specific cell types that express FGF18 have been difficult to identify, and the function of FGF18 in postnatal development and tissue homeostasis has been hampered by the perinatal lethality of Fgf18 null mice. RESULTS: We engineered a floxed allele of Fgf18 (Fgf18 flox ) that allows conditional gene inactivation and a CreERT2 knockin allele (Fgf18 CreERT2 ) that allows the precise identification of cells that express Fgf18 and their lineage. We validated the Fgf18 flox allele by targeting it in mesenchymal tissue and primary mesoderm during embryonic development, resulting in similar phenotypes to those observed in Fgf18 null mice. We also use the Fgf18 CreERT2 allele, in combination with a conditional fluorescent reporter to confirm known and identify new sites of Fgf18 expression. CONCLUSION: These alleles will be useful to investigate FGF18 function during organogenesis and tissue homeostasis, and to target specific cell lineages at embryonic and postnatal time points.


Asunto(s)
Alelos , Factores de Crecimiento de Fibroblastos/metabolismo , Integrasas/genética , Ingeniería de Proteínas/métodos , Animales , Linaje de la Célula , Desarrollo Embrionario , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factores de Crecimiento de Fibroblastos/fisiología , Homeostasis , Mesodermo , Ratones , Organogénesis
12.
Development ; 145(8)2018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29615464

RESUMEN

Embryonic morphogenesis of a complex organism requires proper regulation of patterning and directional growth. Planar cell polarity (PCP) signaling is emerging as a crucial evolutionarily conserved mechanism whereby directional information is conveyed. PCP is thought to be established by global cues, and recent studies have revealed an instructive role of a Wnt signaling gradient in epithelial tissues of both invertebrates and vertebrates. However, it remains unclear whether Wnt/PCP signaling is regulated in a coordinated manner with embryonic patterning during morphogenesis. Here, in mouse developing limbs, we find that apical ectoderm ridge-derived Fgfs required for limb patterning regulate PCP along the proximal-distal axis in a Wnt5a-dependent manner. We demonstrate with genetic evidence that the Wnt5a gradient acts as a global cue that is instructive in establishing PCP in the limb mesenchyme, and that Wnt5a also plays a permissive role to allow Fgf signaling to orient PCP. Our results indicate that limb morphogenesis is regulated by coordination of directional growth and patterning through integration of Wnt5a and Fgf signaling.


Asunto(s)
Tipificación del Cuerpo/fisiología , Polaridad Celular/fisiología , Factor 4 de Crecimiento de Fibroblastos/fisiología , Factor 8 de Crecimiento de Fibroblastos/fisiología , Proteína Wnt-5a/fisiología , Animales , Tipificación del Cuerpo/genética , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Extremidades/embriología , Factor 4 de Crecimiento de Fibroblastos/deficiencia , Factor 4 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/deficiencia , Factor 8 de Crecimiento de Fibroblastos/genética , Mesodermo/embriología , Ratones , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal , Proteína Wnt-5a/deficiencia , Proteína Wnt-5a/genética
13.
Sci Rep ; 7: 43010, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28220837

RESUMEN

Goosecoid (Gsc) expression marks the primary embryonic organizer in vertebrates and beyond. While functions have been assigned during later embryogenesis, the role of Gsc in the organizer has remained enigmatic. Using conditional gain-of-function approaches in Xenopus and mouse to maintain Gsc expression in the organizer and along the axial midline, neural tube closure defects (NTDs) arose and dorsal extension was compromised. Both phenotypes represent convergent extension (CE) defects, arising from impaired Wnt/planar cell polarity (PCP) signaling. Dvl2 recruitment to the cell membrane was inhibited by Gsc in Xenopus animal cap assays and key Wnt/PCP factors (RhoA, Vangl2, Prickle, Wnt11) rescued Gsc-mediated NTDs. Re-evaluation of endogenous Gsc functions in MO-mediated gene knockdown frog and knockout mouse embryos unearthed PCP/CE-related phenotypes as well, including cartilage defects in Xenopus and misalignment of inner ear hair cells in mouse. Our results assign a novel function to Gsc as an inhibitor of Wnt/PCP-mediated CE. We propose that in the organizer Gsc represses CE as well: Gsc-expressing prechordal cells, which leave the organizer first, migrate and do not undergo CE like the Gsc-negative notochordal cells, which subsequently emerge from the organizer. In this model, Gsc provides a switch between cell migration and CE, i.e. cell intercalation.


Asunto(s)
Proteína Goosecoide/metabolismo , Organizadores Embrionarios/metabolismo , Proteínas Wnt/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animales , Polaridad Celular , Proteínas Dishevelled/metabolismo , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Embrión no Mamífero/metabolismo , Embrión no Mamífero/patología , Desarrollo Embrionario , Genes Reporteros , Proteína Goosecoide/deficiencia , Proteína Goosecoide/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Rastreo , Transducción de Señal , Proteínas de Xenopus/genética
14.
Curr Biol ; 26(23): R1237-R1240, 2016 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-27923134

RESUMEN

Two groups have studied the loss of limbs in snake evolution by focusing on a long-distance cis-acting enhancer of Sonic Hedgehog. They find a progressive degeneration of binding sites for key transcription factors, mirroring the progressive limblessness occurring in these reptiles.


Asunto(s)
Evolución Biológica , Serpientes/anatomía & histología , Serpientes/genética , Animales , Peces/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones
15.
PLoS Genet ; 12(5): e1006018, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27144312

RESUMEN

During vertebrate axis extension, adjacent tissue layers undergo profound morphological changes: within the neuroepithelium, neural tube closure and neural crest formation are occurring, while within the paraxial mesoderm somites are segmenting from the presomitic mesoderm (PSM). Little is known about the signals between these tissues that regulate their coordinated morphogenesis. Here, we analyze the posterior axis truncation of mouse Fgf3 null homozygotes and demonstrate that the earliest role of PSM-derived FGF3 is to regulate BMP signals in the adjacent neuroepithelium. FGF3 loss causes elevated BMP signals leading to increased neuroepithelium proliferation, delay in neural tube closure and premature neural crest specification. We demonstrate that elevated BMP4 depletes PSM progenitors in vitro, phenocopying the Fgf3 mutant, suggesting that excessive BMP signals cause the Fgf3 axis defect. To test this in vivo we increased BMP signaling in Fgf3 mutants by removing one copy of Noggin, which encodes a BMP antagonist. In such mutants, all parameters of the Fgf3 phenotype were exacerbated: neural tube closure delay, premature neural crest specification, and premature axis termination. Conversely, genetically decreasing BMP signaling in Fgf3 mutants, via loss of BMP receptor activity, alleviates morphological defects. Aberrant apoptosis is observed in the Fgf3 mutant tailbud. However, we demonstrate that cell death does not cause the Fgf3 phenotype: blocking apoptosis via deletion of pro-apoptotic genes surprisingly increases all Fgf3 defects including causing spina bifida. We demonstrate that this counterintuitive consequence of blocking apoptosis is caused by the increased survival of BMP-producing cells in the neuroepithelium. Thus, we show that FGF3 in the caudal vertebrate embryo regulates BMP signaling in the neuroepithelium, which in turn regulates neural tube closure, neural crest specification and axis termination. Uncovering this FGF3-BMP signaling axis is a major advance toward understanding how these tissue layers interact during axis extension with important implications in human disease.


Asunto(s)
Proteína Morfogenética Ósea 4/genética , Proteínas Portadoras/genética , Factor 3 de Crecimiento de Fibroblastos/genética , Cresta Neural/metabolismo , Defectos del Tubo Neural/genética , Animales , Tipificación del Cuerpo/genética , Proteína Morfogenética Ósea 4/metabolismo , Proteínas Portadoras/metabolismo , Muerte Celular/genética , Factor 3 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Ratones , Cresta Neural/crecimiento & desarrollo , Tubo Neural/crecimiento & desarrollo , Tubo Neural/metabolismo , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/patología , Transducción de Señal/genética , Somitos/crecimiento & desarrollo , Somitos/metabolismo
16.
Dev Biol ; 411(1): 72-84, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26794256

RESUMEN

Fibroblast growth factor (FGF) signaling is a critical regulator of skeletal development. Fgf9 and Fgf18 are the only FGF ligands with identified functions in embryonic bone growth. Mice lacking Fgf9 or Fgf18 have distinct skeletal phenotypes; however, the extent of overlapping or redundant functions for these ligands and the stage-specific contributions of FGF signaling to chondrogenesis and osteogenesis are not known. To identify separate versus shared roles for FGF9 and FGF18, we generated a combined series of Fgf9 and Fgf18 null alleles. Analysis of embryos lacking alleles of Fgf9 and Fgf18 shows that both encoded ligands function redundantly to control all stages of skeletogenesis; however, they have variable potencies along the proximodistal limb axis, suggesting gradients of activity during formation of the appendicular skeleton. Congenital absence of both Fgf9 and Fgf18 results in a striking osteochondrodysplasia and revealed functions for FGF signaling in early proximal limb chondrogenesis. Additional defects were also noted in craniofacial bones, vertebrae, and ribs. Loss of alleles of Fgf9 and Fgf18 also affect the expression of genes encoding other key intrinsic skeletal regulators, including IHH, PTHLH (PTHrP), and RUNX2, revealing potential direct, indirect, and compensatory mechanisms to coordinate chondrogenesis and osteogenesis.


Asunto(s)
Desarrollo Óseo/genética , Huesos/embriología , Condrogénesis/genética , Factor 9 de Crecimiento de Fibroblastos/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Osteocondrodisplasias/genética , Osteogénesis/genética , Animales , Huesos/anomalías , Diferenciación Celular , Condrocitos/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Factor 9 de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/genética , Placa de Crecimiento/embriología , Proteínas Hedgehog/biosíntesis , Ratones , Ratones Noqueados , Proteína Relacionada con la Hormona Paratiroidea/biosíntesis , Transducción de Señal/genética
17.
Dev Biol ; 411(2): 266-276, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26826495

RESUMEN

During vertebrate embryogenesis the interdigital mesenchyme is removed by programmed cell death (PCD), except in species with webbed limbs. Although bone morphogenetic proteins (BMPs) have long been known to be players in this process, it is unclear if they play a direct role in the interdigital mesenchyme or if they only act indirectly, by affecting fibroblast growth factor (FGF) signaling. A series of genetic studies have shown that BMPs act indirectly by regulating the withdrawal of FGF activity from the apical ectodermal ridge (AER); this FGF activity acts as a cell survival factor for the underlying mesenchyme. Other studies using exogenous factors to inhibit BMP activity in explanted mouse limbs suggest that BMPs do not act directly in the mesenchyme. To address the question of whether BMPs act directly, we used an interdigit-specific Cre line to inactivate several genes that encode components of the BMP signaling pathway, without perturbing the normal downregulation of AER-FGF activity. Of three Bmps expressed in the interdigital mesenchyme, Bmp7 is necessary for PCD, but Bmp2 and Bmp4 both have redundant roles, with Bmp2 being the more prominent player. Removing BMP signals to the interdigit by deleting the receptor gene, Bmpr1a, causes a loss of PCD and syndactyly, thereby unequivocally proving that BMPs are direct triggers of PCD in this tissue. We present a model in which two events must occur for normal interdigital PCD: the presence of a BMP death trigger and the absence of an FGF survival activity. We demonstrate that neither event is required for formation of the interdigital vasculature, which is necessary for PCD. However, both events converge on the production of reactive oxygen species that activate PCD.


Asunto(s)
Apoptosis , Proteína Morfogenética Ósea 2/fisiología , Proteína Morfogenética Ósea 4/fisiología , Proteína Morfogenética Ósea 7/fisiología , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Regulación del Desarrollo de la Expresión Génica , Animales , Cruzamientos Genéticos , Extremidades/embriología , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Miembro Anterior/patología , Integrasas/metabolismo , Masculino , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Sindactilia/genética , Factores de Tiempo , Dedos del Pie/patología , beta-Galactosidasa/metabolismo
18.
Dev Neurobiol ; 76(9): 956-71, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26600420

RESUMEN

A prevalent developmental mechanism for the assignment of cell identities is the production of spatiotemporal concentration gradients of extracellular signaling molecules that are interpreted by the responding cells. One of such signaling systems is the Shh gradient that controls neuronal subtype identity in the ventral spinal cord. Using loss and gain of function approaches in chick and mouse embryos, we show here that the fibroblast growth factor (FGF) signaling pathway is required to restrict the domains of ventral gene expression as neuroepithelial cells become exposed to Shh during caudal extension of the embryo. FGF signaling activates the expression of the Shh receptor and negative pathway regulator Patched 2 (Ptch2) and therefore can enhance a negative feedback loop that restrains the activity of the pathway. Thus, we identify one of the mechanisms by which FGF signaling acts as a modulator of the onset of Shh signaling activity in the context of coordination of ventral patterning and caudal axis extension. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 76: 956-971, 2016.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Hedgehog/fisiología , Transducción de Señal/fisiología , Asta Ventral de la Médula Espinal/fisiología , Animales , Embrión de Pollo , Humanos , Ratones , Asta Ventral de la Médula Espinal/embriología
19.
Genesis ; 54(2): 91-8, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26666435

RESUMEN

The fibroblast growth factor (FGF) family consists of 22 ligands in mice and humans. FGF signaling is vital for embryogenesis and, when dysregulated, can cause disease. Loss-of-function genetic analysis in the mouse has been crucial for understanding FGF function. Such analysis has revealed that multiple Fgfs sometimes function redundantly. Exploring such redundancy between Fgf3 and Fgf4 is currently impossible because both genes are located on chromosome 7, about 18.5 kb apart, making the frequency of interallelic cross-over between existing mutant alleles too infrequent to be practicable. Therefore, we retargeted Fgf3 and Fgf4 in cis, generating an Fgf3 null allele and a conditional Fgf4 allele, subject to Cre inactivation. To increase the frequency of cis targeting, we used an F1 embryonic stem cell line that contained 129/SvJae (129) and C57BL/6J (B6) chromosomes and targeting constructs isogenic to the 129 chromosome. We confirmed cis targeting by assaying for B6/129 allele-specific single-nucleotide polymorphisms. We demonstrated the utility of the Fgf3(Δ)-Fgf4(flox)-cis mouse line by showing that the caudal axis extension defects found in the Fgf3 mutants worsen when Fgf4 is also inactivated. This Fgf3(Δ)-Fgf4(flox)-cis line will be useful to study redundancy of these genes in a variety of tissues and stages in development.


Asunto(s)
Línea Celular , Factor 3 de Crecimiento de Fibroblastos/genética , Factor 4 de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/fisiología , Animales , Células Madre Embrionarias , Marcación de Gen , Ratones , Ratones Endogámicos C57BL
20.
Dev Cell ; 35(3): 322-32, 2015 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-26555052

RESUMEN

The mammalian lung forms its elaborate tree-like structure following a largely stereotypical branching sequence. While a number of genes have been identified to play essential roles in lung branching, what coordinates the choice between branch growth and new branch formation has not been elucidated. Here we show that loss of FGF-activated transcription factor genes, Etv4 and Etv5 (collectively Etv), led to prolonged branch tip growth and delayed new branch formation. Unexpectedly, this phenotype is more similar to mutants with increased rather than decreased FGF activity. Indeed, an increased Fgf10 expression is observed, and reducing Fgf10 dosage can attenuate the Etv mutant phenotype. Further evidence indicates that ETV inhibits Fgf10 via directly promoting Shh expression. SHH in turn inhibits local Fgf10 expression and redirects growth, thereby initiating new branches. Together, our findings establish ETV as a key node in the FGF-ETV-SHH inhibitory feedback loop that dictates branching periodicity.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Pulmón/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Factores de Transcripción/metabolismo , Animales , Tipificación del Cuerpo/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Proteínas Hedgehog/metabolismo , Ratones , Ratones Transgénicos , Morfogénesis/genética , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...