Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Genes Dev ; 38(9-10): 393-414, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38834239

RESUMEN

The fibroblast growth factor (FGF) pathway is a conserved signaling pathway required for embryonic development. Activated FGF receptor 1 (FGFR1) drives multiple intracellular signaling cascade pathways, including ERK/MAPK and PI3K/AKT, collectively termed canonical signaling. However, unlike Fgfr1-null embryos, embryos containing hypomorphic mutations in Fgfr1 lacking the ability to activate canonical downstream signals are still able to develop to birth but exhibit severe defects in all mesodermal-derived tissues. The introduction of an additional signaling mutation further reduces the activity of Fgfr1, leading to earlier lethality, reduced somitogenesis, and more severe changes in transcriptional outputs. Genes involved in migration, ECM interaction, and phosphoinositol signaling were significantly downregulated, proteomic analysis identified changes in interactions with endocytic pathway components, and cells expressing mutant receptors show changes in endocytic trafficking. Together, we identified processes regulating early mesoderm development by mechanisms involving both canonical and noncanonical Fgfr1 pathways, including direct interaction with cell adhesion components and endocytic regulation.


Asunto(s)
Endocitosis , Regulación del Desarrollo de la Expresión Génica , Mesodermo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Transducción de Señal , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Animales , Mesodermo/embriología , Mesodermo/metabolismo , Transducción de Señal/genética , Endocitosis/genética , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Desarrollo Embrionario/genética , Transporte de Proteínas , Mutación
2.
bioRxiv ; 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38405698

RESUMEN

The Fibroblast growth factor (FGF) pathway is a conserved signaling pathway required for embryonic development. Activated FGF receptor 1 (FGFR1) drives multiple intracellular signaling cascade pathways, including ERK/MAPK and PI3K/AKT, collectively termed canonical signaling. However, unlike Fgfr1 null embryos, embryos containing hypomorphic mutations in Fgfr1 lacking the ability to activate canonical downstream signals are still able to develop to birth, but exhibit severe defects in all mesodermal-derived tissues. The introduction of an additional signaling mutation further reduces the activity of Fgfr1, leading to earlier lethality, reduced somitogenesis, and more severe changes in transcriptional outputs. Genes involved in migration, ECM-interaction, and phosphoinositol signaling were significantly downregulated, proteomic analysis identified changes in interactions with endocytic pathway components, and cells expressing mutant receptors show changes in endocytic trafficking. Together, we identify processes regulating early mesoderm development by mechanisms involving both canonical and non-canonical Fgfr1 pathways, including direct interaction with cell adhesion components and endocytic regulation.

3.
Dev Biol ; 503: 113-119, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37660778

RESUMEN

Understanding how cells activate intracellular signaling pathways in response to external signals, such as growth factors, is a longstanding goal of cell and developmental biology. Recently, live-cell signaling reporters have greatly expanded our understanding of signaling dynamics in response to wide-ranging stimuli and chemical or genetic perturbation, both ex vivo (cell lines) and in vivo (whole embryos or animals). Among the many varieties of reporter systems, translocation reporters that change sub-cellular localization in response to pathway activation have received considerable attention for their ease of use compared to FRET systems and favorable response times compared to transcriptional reporters. We reasoned that mouse reporter lines expressed in a conditional fashion would be a useful addition to the arsenal of mouse genetic tools, as such lines remain undeveloped despite widespread use of these sensors. We therefore created and validated two novel mouse reporter lines at the ROSA26 locus. One expresses an ERK1/2 pathway reporter and a nuclear marker from a single transcript, while the second additionally expresses an AKT reporter in order to simultaneously interrogate both pathways.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Proteínas Proto-Oncogénicas c-akt , Animales , Ratones , Línea Celular , Embrión de Mamíferos , Transducción de Señal , Colorantes Fluorescentes/metabolismo , Transporte de Proteínas
4.
Biol Open ; 12(7)2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37421147

RESUMEN

FGF activation is known to engage canonical signals, including ERK/MAPK and PI3K/AKT, through various effectors including FRS2 and GRB2. Fgfr2FCPG/FCPG mutants that abrogate canonical intracellular signaling exhibit a range of mild phenotypes but are viable, in contrast to embryonic lethal Fgfr2-/- mutants. GRB2 has been reported to interact with FGFR2 through a non-traditional mechanism, by binding to the C-terminus of FGFR2 independently of FRS2 recruitment. To investigate whether this interaction provides functionality beyond canonical signaling, we generated mutant mice harboring a C-terminal truncation (T). We found that Fgfr2T/T mice are viable and have no distinguishable phenotype, indicating that GRB2 binding to the C-terminal end of FGFR2 is not required for development or adult homeostasis. We further introduced the T mutation on the sensitized FCPG background but found that Fgfr2FCPGT/FCPGT mutants did not exhibit significantly more severe phenotypes. We therefore conclude that, although GRB2 can bind to FGFR2 independently of FRS2, this binding does not have a critical role in development or homeostasis.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Transducción de Señal , Animales , Ratones , Desarrollo Embrionario/genética , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Mutación , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/genética
5.
Development ; 150(6)2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36861436

RESUMEN

Loss of FGF signaling leads to defects in salivary gland branching, but the mechanisms underlying this phenotype remain largely unknown. We disrupted expression of Fgfr1 and Fgfr2 in salivary gland epithelial cells and found that both receptors function coordinately in regulating branching. Strikingly, branching morphogenesis in double knockouts is restored by Fgfr1 and Fgfr2 (Fgfr1/2) knock-in alleles incapable of engaging canonical RTK signaling, suggesting that additional FGF-dependent mechanisms play a role in salivary gland branching. Fgfr1/2 conditional null mutants showed defective cell-cell and cell-matrix adhesion, both of which have been shown to play instructive roles in salivary gland branching. Loss of FGF signaling led to disordered cell-basement membrane interactions in vivo as well as in organ culture. This was partially restored upon introducing Fgfr1/2 wild-type or signaling alleles that are incapable of eliciting canonical intracellular signaling. Together, our results identify non-canonical FGF signaling mechanisms that regulate branching morphogenesis through cell-adhesion processes.


Asunto(s)
Células Epiteliales , Transducción de Señal , Adhesión Celular/genética , Células Epiteliales/metabolismo , Morfogénesis/genética , Glándulas Salivales , Transducción de Señal/genética , Factores de Crecimiento de Fibroblastos
6.
bioRxiv ; 2023 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-36993499

RESUMEN

FGF activation is known to engage canonical signals, including ERK/MAPK and PI3K/AKT, through various effectors including FRS2 and GRB2. Fgfr2 FCPG/FCPG mutants that abrogate canonical intracellular signaling exhibit a range of mild phenotypes but are viable in contrast to embryonic lethal Fgfr2 -/- mutants. GRB2 has been reported to interact with FGFR2 through a non-traditional mechanism, by binding to the C-terminus of FGFR2 independently of FRS2 recruitment. To investigate if this interaction provides functionality beyond canonical signaling, we generated mutant mice harboring a C-terminal truncation (T). We found that Fgfr2 T/T mice are viable and have no distinguishable phenotype, indicating that GRB2 binding to the C-terminal end of FGFR2 is not required for development or adult homeostasis. We further introduced the T mutation on the sensitized FCPG background but found that Fgfr2 FCPGT/FCPGT mutants did not exhibit significantly more severe phenotypes. We therefore conclude that, while GRB2 can bind to FGFR2 independently of FRS2, this binding does not have a critical role in development or homeostasis.

7.
Curr Top Dev Biol ; 149: 123-152, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35606055

RESUMEN

Receptor tyrosine kinases (RTKs) are a conserved superfamily of transmembrane growth factor receptors that drive numerous cellular processes during development and in the adult. Upon activation, multiple adaptors and signaling effector proteins are recruited to binding site motifs located within the intracellular domain of the RTK. These RTK-effector interactions drive subsequent intracellular signaling cascades involved in canonical RTK signaling. Genetic dissection has revealed that alleles of Fibroblast Growth Factor receptors (FGFRs) that lack all canonical RTK signaling still retain some kinase-dependent biological activity. Here we examine how genetic analysis can be used to understand the mechanism by which RTKs drive multiple developmental processes via canonical signaling while revealing noncanonical activities. Recent data from both FGFRs and other RTKs highlight potential noncanonical roles in cell adhesion and nuclear signaling. The data supporting such functions are discussed as are recent technologies that have the potential to provide valuable insight into the developmental significance of these noncanonical activities.


Asunto(s)
Proteínas Tirosina Quinasas Receptoras , Transducción de Señal , Fosforilación , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/fisiología , Tirosina/metabolismo
8.
Curr Top Dev Biol ; 149: xi-xii, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35606064
9.
Genesis ; 60(3): e23468, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35180326

RESUMEN

The Wnt1-Cre transgenic mouse line is widely used to express the CRE recombinase in neural crest lineages, but it overexpresses WNT1 itself, which can cause undesired phenotypes. To address this, we and others previously developed a Wnt1-Cre2 line based on the same regulatory elements as Wnt1-Cre but without ectopic Wnt1 expression. However, while Wnt1-Cre2 exhibits normal activity when transmitted from female mice, it exhibits unexpected activity in the male germline. The Wnt1-Cre2 transgene was previously mapped to the E2f1 locus. Several genes in this genomic region exhibit significant expression in spermatogonia or spermatocytes, suggesting that local regulatory elements may be driving ectopic transgene expression. The Wnt1-Cre2 line can therefore be used both as a neural crest specific and a general deleter, and care should be taken when setting up genetic crosses.


Asunto(s)
Integrasas , Cresta Neural , Animales , Femenino , Células Germinativas/metabolismo , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Cresta Neural/metabolismo , Fenotipo , Transgenes
10.
Elife ; 112022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-35044299

RESUMEN

Serum response factor (SRF) is an essential transcription factor that influences many cellular processes including cell proliferation, migration, and differentiation. SRF directly regulates and is required for immediate early gene (IEG) and actin cytoskeleton-related gene expression. SRF coordinates these competing transcription programs through discrete sets of cofactors, the ternary complex factors (TCFs) and myocardin-related transcription factors (MRTFs). The relative contribution of these two programs to in vivo SRF activity and mutant phenotypes is not fully understood. To study how SRF utilizes its cofactors during development, we generated a knock-in SrfaI allele in mice harboring point mutations that disrupt SRF-MRTF-DNA complex formation but leave SRF-TCF activity unaffected. Homozygous SrfaI/aI mutants die at E10.5 with notable cardiovascular phenotypes, and neural crest conditional mutants succumb at birth to defects of the cardiac outflow tract but display none of the craniofacial phenotypes associated with complete loss of SRF in that lineage. Our studies further support an important role for MRTF mediating SRF function in cardiac neural crest and suggest new mechanisms by which SRF regulates transcription during development.


Asunto(s)
Cresta Neural/embriología , Factor de Respuesta Sérica/genética , Factores Complejos Ternarios/genética , Factores de Transcripción/genética , Animales , Ratones , Factor de Respuesta Sérica/metabolismo , Factores Complejos Ternarios/metabolismo , Factores de Transcripción/metabolismo
12.
Genes Dev ; 34(23-24): 1735-1752, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33184218

RESUMEN

FGFs are key developmental regulators that engage a signal transduction cascade through receptor tyrosine kinases, prominently engaging ERK1/2 but also other pathways. However, it remains unknown whether all FGF activities depend on this canonical signal transduction cascade. To address this question, we generated allelic series of knock-in Fgfr1 and Fgfr2 mouse strains, carrying point mutations that disrupt binding of signaling effectors, and a kinase dead allele of Fgfr2 that broadly phenocopies the null mutant. When interrogated in cranial neural crest cells, we identified discrete functions for signaling pathways in specific craniofacial contexts, but point mutations, even when combined, failed to recapitulate the single or double null mutant phenotypes. Furthermore, the signaling mutations abrogated established FGF-induced signal transduction pathways, yet FGF functions such as cell-matrix and cell-cell adhesion remained unaffected, though these activities did require FGFR kinase activity. Our studies establish combinatorial roles of Fgfr1 and Fgfr2 in development and uncouple novel FGFR kinase-dependent cell adhesion properties from canonical intracellular signaling.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Transducción de Señal/genética , Animales , Adhesión Celular/genética , Muerte Celular/genética , Células Cultivadas , Ratones , Mutación , Cresta Neural/citología , Proteínas Quinasas/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo
13.
Genes Dev ; 34(19-20): 1256-1286, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33004485

RESUMEN

The mouse is one of the most widely used model organisms for genetic study. The tools available to alter the mouse genome have developed over the preceding decades from forward screens to gene targeting in stem cells to the recent influx of CRISPR approaches. In this review, we first consider the history of mice in genetic study, the development of classic approaches to genome modification, and how such approaches have been used and improved in recent years. We then turn to the recent surge of nuclease-mediated techniques and how they are changing the field of mouse genetics. Finally, we survey common classes of alleles used in mice and discuss how they might be engineered using different methods.


Asunto(s)
Técnicas Genéticas/tendencias , Ratones/genética , Modelos Animales , Animales
14.
Dev Cell ; 53(3): 255-256, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32369738

RESUMEN

Neural crest cells have different developmental potencies at different levels along the body axis of the embryo. In this issue of Developmental Cell, Gandhi et al. identify transcription factors that define one subtype of neural crest, the cardiac crest, and demonstrate their ability to reprogram trunk into cardiac crest.


Asunto(s)
Cardiopatías Congénitas , Cresta Neural , Corazón , Proteínas de Homeodominio , Humanos , Proteínas Represoras , Factores de Transcripción
15.
Dev Biol ; 447(2): 127-136, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30905353
16.
Dev Biol ; 446(1): 94-101, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30552867

RESUMEN

FGF signaling plays important roles in many aspects of mammalian development. Fgfr1-/- and Fgfr1-/-Fgfr2-/- mouse embryos on a 129S4 co-isogenic background fail to survive past the peri-implantation stage, whereas Fgfr2-/- embryos die at midgestation and show defects in limb and placental development. To investigate the basis for the Fgfr1-/- and Fgfr1-/-Fgfr2-/- peri-implantation lethality, we examined the role of FGFR1 and FGFR2 in trophectoderm (TE) development. In vivo, Fgfr1-/- TE cells failed to downregulate CDX2 in the mural compartment and exhibited abnormal apicobasal E-Cadherin polarity. In vitro, we were able to derive mutant trophoblast stem cells (TSCs) from Fgfr1-/- or Fgfr2-/- single mutant, but not from Fgfr1-/-Fgfr2-/- double mutant blastocysts. Fgfr1-/- TSCs however failed to efficiently upregulate TE differentiation markers upon differentiation. These results suggest that while the TE is specified in Fgfr1-/- mutants, its differentiation abilities are compromised leading to defects at implantation.


Asunto(s)
Implantación del Embrión/genética , Regulación del Desarrollo de la Expresión Génica , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Trofoblastos/metabolismo , Animales , Factor de Transcripción CDX2/genética , Factor de Transcripción CDX2/metabolismo , Diferenciación Celular/genética , Línea Celular , Células Cultivadas , Ectodermo/citología , Femenino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Embarazo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Trofoblastos/citología
17.
Dev Biol ; 442(1): 155-161, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30026121

RESUMEN

FGF signaling is known to play a critical role in the specification of primitive endoderm (PrE) and epiblast (Epi) from the inner cell mass (ICM) during mouse preimplantation development, but how FGFs synergize with other growth factor signaling pathways is unknown. Because PDGFRα signaling has also been implicated in the PrE, we investigated the coordinate functions of PDGFRα together with FGFR1 or FGFR2 in PrE development. PrE development was abrogated in Pdgfra; Fgfr1 compound mutants, or significantly reduced in Pdgfra; Fgfr2 or PdgfraPI3K; Fgfr2 compound mutants. We provide evidence that both Fgfr2 and Pdgfra play roles in PrE cell survival while Fgfr1 controls PrE cell specification. Our results suggest a model where FGFR1-engaged ERK1/2 signaling governs PrE specification while PDGFRα- and by analogy possibly FGFR2- engaged PI3K signaling regulates PrE survival and positioning in the embryo. Together, these studies indicate how multiple growth factors and signaling pathways can cooperate in preimplantation development.


Asunto(s)
Factor 4 de Crecimiento de Fibroblastos/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Blastocisto/metabolismo , Masa Celular Interna del Blastocisto/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Endodermo/metabolismo , Factor 4 de Crecimiento de Fibroblastos/fisiología , Factores de Crecimiento de Fibroblastos/metabolismo , Estratos Germinativos/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología
19.
Dev Biol ; 444 Suppl 1: S79-S97, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29453943

RESUMEN

Receptor tyrosine kinase-mediated growth factor signaling is essential for proper formation and development of the neural crest. The many ligands and receptors implicated in these processes signal through relatively few downstream pathways, frequently converging on the MAPK and PI3K pathways. Despite decades of study, there is still considerable uncertainty about where and when these signaling pathways are required and how they elicit particular responses. This review summarizes our current understanding of growth factor-induced MAPK and PI3K signaling in the neural crest.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Cresta Neural/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Evolución Biológica , Tipificación del Cuerpo/fisiología , Diferenciación Celular , Movimiento Celular , Receptores ErbB/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Cresta Neural/embriología , Neurogénesis , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Vertebrados/embriología
20.
Development ; 144(21): 4026-4036, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28947535

RESUMEN

Craniosynostosis is a prevalent human birth defect characterized by premature fusion of calvarial bones. In this study, we show that tight regulation of endogenous PDGFRα activity is required for normal calvarium development in the mouse and that dysregulated PDGFRα activity causes craniosynostosis. Constitutive activation of PDGFRα leads to expansion of cartilage underlying the coronal sutures, which contribute to suture closure through endochondral ossification, in a process regulated in part by PI3K/AKT signaling. Our results thus identify a novel mechanism underlying calvarial development in craniosynostosis.


Asunto(s)
Cartílago/embriología , Suturas Craneales/embriología , Suturas Craneales/metabolismo , Craneosinostosis/metabolismo , Morfogénesis , Osteogénesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Alelos , Animales , Cartílago/anomalías , Cartílago/metabolismo , Linaje de la Célula , Condrogénesis , Suturas Craneales/patología , Regulación del Desarrollo de la Expresión Génica , Ligandos , Mesodermo/metabolismo , Ratones Endogámicos C57BL , Cresta Neural/metabolismo , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal , Cráneo/anomalías , Cráneo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...