Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 967
Filtrar
1.
Dev Biol ; 516: 59-70, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39098630

RESUMEN

Dicer substrate interfering RNAs (DsiRNAs) destroy targeted transcripts using the RNA-Induced Silencing Complex (RISC) through a process called RNA interference (RNAi). This process is ubiquitous among eukaryotes. Here we report the utility of DsiRNA in embryos of the sea urchin Lytechinus variegatus (Lv). Specific knockdowns phenocopy known morpholino and inhibitor knockdowns, and DsiRNA offers a useful alternative to morpholinos. Methods are described for the design of specific DsiRNAs that lead to destruction of targeted mRNA. DsiRNAs directed against pks1, an enzyme necessary for pigment production, show how successful DsiRNA perturbations are monitored by RNA in situ analysis and by qPCR to determine relative destruction of targeted mRNA. DsiRNA-based knockdowns phenocopy morpholino- and drug-based inhibition of nodal and lefty. Other knockdowns demonstrate that the RISC operates early in development as well as on genes that are first transcribed hours after gastrulation is completed. Thus, DsiRNAs effectively mediate destruction of targeted mRNA in the sea urchin embryo. The approach offers significant advantages over other widely used methods in the urchin in terms of cost, and ease of procurement, and offers sizeable experimental advantages in terms of ease of handling, injection, and knockdown validation.


Asunto(s)
Técnicas de Silenciamiento del Gen , Proteína Nodal , Interferencia de ARN , Transducción de Señal , Animales , Proteína Nodal/metabolismo , Proteína Nodal/genética , Transducción de Señal/genética , Técnicas de Silenciamiento del Gen/métodos , Erizos de Mar/genética , Erizos de Mar/embriología , Regulación del Desarrollo de la Expresión Génica , Morfolinos/genética , Morfolinos/farmacología , Embrión no Mamífero/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Lytechinus/genética , Lytechinus/embriología , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/genética , Ribonucleasa III/metabolismo , Ribonucleasa III/genética
2.
Stem Cell Reports ; 19(9): 1320-1335, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39151429

RESUMEN

The homeostasis of human pluripotent stem cells (hPSCs) requires the signaling balance of extracellular factors. Exogenous regulators from cell culture medium have been widely reported, but little attention has been paid to the autocrine factor from hPSCs themselves. In this report, we demonstrate that extracellular signal-related kinase 5 (ERK5) regulates endogenous autocrine factors essential for pluripotency and differentiation. ERK5 inhibition leads to erroneous cell fate specification in all lineages even under lineage-specific induction. hPSCs can self-renew under ERK5 inhibition in the presence of fibroblast growth factor 2 (FGF2) and transforming growth factor ß (TGF-ß), although NANOG expression is partially suppressed. Further analysis demonstrates that ERK5 promotes the expression of autocrine factors such as NODAL, FGF8, and WNT3. The addition of NODAL protein rescues NANOG expression and differentiation phenotypes under ERK5 inhibition. We demonstrate that constitutively active ERK5 pathway allows self-renewal even without essential growth factors FGF2 and TGF-ß. This study highlights the essential contribution of autocrine pathways to proper maintenance and differentiation.


Asunto(s)
Comunicación Autocrina , Proteína Quinasa 7 Activada por Mitógenos , Proteína Homeótica Nanog , Células Madre Pluripotentes , Humanos , Diferenciación Celular , Línea Celular , Linaje de la Célula , Proliferación Celular , Autorrenovación de las Células , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/genética , Proteína Homeótica Nanog/metabolismo , Proteína Homeótica Nanog/genética , Proteína Nodal/metabolismo , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/citología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
3.
Genome Med ; 16(1): 100, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39138574

RESUMEN

To date, the role of NODAL in normal and abnormal L-R asymmetry has been well established. In a recent paper, mutations of this gene have been reported in heterotaxy but also in transposition with D- or L-ventricular loop. The effects of NODAL and other laterality genes can be recognized separately in all three cardiac segments: for topology and septation of the atria, for ventricular looping, and for spiralization and alignment of the great arteries.


Asunto(s)
Cardiopatías Congénitas , Síndrome de Heterotaxia , Humanos , Síndrome de Heterotaxia/genética , Cardiopatías Congénitas/genética , Proteína Nodal/genética , Proteína Nodal/metabolismo , Corazón , Mutación , Animales
4.
Nat Commun ; 15(1): 6547, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39095343

RESUMEN

Myosin1D (Myo1D) has recently emerged as a conserved regulator of animal Left-Right (LR) asymmetry that governs the morphogenesis of the vertebrate central LR Organizer (LRO). In addition to Myo1D, the zebrafish genome encodes the closely related Myo1G. Here we show that while Myo1G also controls LR asymmetry, it does so through an entirely different mechanism. Myo1G promotes the Nodal-mediated transfer of laterality information from the LRO to target tissues. At the cellular level, Myo1G is associated with endosomes positive for the TGFß signaling adapter SARA. myo1g mutants have fewer SARA-positive Activin receptor endosomes and a reduced responsiveness to Nodal ligands that results in a delay of left-sided Nodal propagation and tissue-specific laterality defects in organs that are most distant from the LRO. Additionally, Myo1G promotes signaling by different Nodal ligands in specific biological contexts. Our findings therefore identify Myo1G as a context-dependent regulator of the Nodal signaling pathway.


Asunto(s)
Tipificación del Cuerpo , Transducción de Señal , Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/genética , Pez Cebra/metabolismo , Pez Cebra/embriología , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Tipificación del Cuerpo/genética , Proteína Nodal/metabolismo , Proteína Nodal/genética , Regulación del Desarrollo de la Expresión Génica , Endosomas/metabolismo , Miosinas/metabolismo , Miosinas/genética , Mutación , Miosina Tipo I/metabolismo , Miosina Tipo I/genética , Embrión no Mamífero/metabolismo
5.
Proc Natl Acad Sci U S A ; 121(28): e2408346121, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38968117

RESUMEN

Xenopus embryos provide a favorable material to dissect the sequential steps that lead to dorsal-ventral (D-V) and anterior-posterior (A-P) cell differentiation. Here, we analyze the signaling pathways involved in this process using loss-of-function and gain-of-function approaches. The initial step was provided by Hwa, a transmembrane protein that robustly activates early ß-catenin signaling when microinjected into the ventral side of the embryo leading to complete twinned axes. The following step was the activation of Xenopus Nodal-related growth factors, which could rescue the depletion of ß-catenin and were themselves blocked by the extracellular Nodal antagonists Cerberus-Short and Lefty. During gastrulation, the Spemann-Mangold organizer secretes a cocktail of growth factor antagonists, of which the BMP antagonists Chordin and Noggin could rescue simultaneously D-V and A-P tissues in ß-catenin-depleted embryos. Surprisingly, this rescue occurred in the absence of any ß-catenin transcriptional activity as measured by ß-catenin activated Luciferase reporters. The Wnt antagonist Dickkopf (Dkk1) strongly synergized with the early Hwa signal by inhibiting late Wnt signals. Depletion of Sizzled (Szl), an antagonist of the Tolloid chordinase, was epistatic over the Hwa and Dkk1 synergy. BMP4 mRNA injection blocked Hwa-induced ectopic axes, and Dkk1 inhibited BMP signaling late, but not early, during gastrulation. Several unexpected findings were made, e.g., well-patterned complete embryonic axes are induced by Chordin or Nodal in ß-catenin knockdown embryos, dorsalization by Lithium chloride (LiCl) is mediated by Nodals, Dkk1 exerts its anteriorizing and dorsalizing effects by regulating late BMP signaling, and the Dkk1 phenotype requires Szl.


Asunto(s)
Tipificación del Cuerpo , Péptidos y Proteínas de Señalización Intercelular , Transducción de Señal , Proteínas de Xenopus , beta Catenina , Animales , Tipificación del Cuerpo/genética , Proteínas de Xenopus/metabolismo , Proteínas de Xenopus/genética , beta Catenina/metabolismo , beta Catenina/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Xenopus laevis/embriología , Regulación del Desarrollo de la Expresión Génica , Gastrulación , Proteína Nodal/metabolismo , Proteína Nodal/genética , Embrión no Mamífero/metabolismo , Embrión no Mamífero/embriología , Organizadores Embrionarios/metabolismo , Glicoproteínas
6.
Adv Exp Med Biol ; 1441: 167-183, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884711

RESUMEN

Formation of the vertebrate heart with its complex arterial and venous connections is critically dependent on patterning of the left-right axis during early embryonic development. Abnormalities in left-right patterning can lead to a variety of complex life-threatening congenital heart defects. A highly conserved pathway responsible for left-right axis specification has been uncovered. This pathway involves initial asymmetric activation of a nodal signaling cascade at the embryonic node, followed by its propagation to the left lateral plate mesoderm and activation of left-sided expression of the Pitx2 transcription factor specifying visceral organ asymmetry. Intriguingly, recent work suggests that cardiac laterality is encoded by intrinsic cell and tissue chirality independent of Nodal signaling. Thus, Nodal signaling may be superimposed on this intrinsic chirality, providing additional instructive cues to pattern cardiac situs. The impact of intrinsic chirality and the perturbation of left-right patterning on myofiber organization and cardiac function warrants further investigation. We summarize recent insights gained from studies in animal models and also some human clinical studies in a brief overview of the complex processes regulating cardiac asymmetry and their impact on cardiac function and the pathogenesis of congenital heart defects.


Asunto(s)
Tipificación del Cuerpo , Cardiopatías Congénitas , Corazón , Humanos , Animales , Corazón/embriología , Corazón/fisiología , Tipificación del Cuerpo/genética , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/fisiopatología , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/patología , Transducción de Señal , Regulación del Desarrollo de la Expresión Génica , Proteína Nodal/metabolismo , Proteína Nodal/genética
7.
Science ; 384(6700): 1105-1110, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38843334

RESUMEN

Axis formation in fish and amphibians typically begins with a prepattern of maternal gene products. Annual killifish embryogenesis, however, challenges prepatterning models as blastomeres disperse and then aggregate to form the germ layers and body axes. We show that huluwa, a prepatterning factor thought to break symmetry by stabilizing ß-catenin, is truncated and inactive in Nothobranchius furzeri. Nuclear ß-catenin is not selectively stabilized on one side of the blastula but accumulates in cells forming the aggregate. Blocking ß-catenin activity or Nodal signaling disrupts aggregate formation and germ layer specification. Nodal signaling coordinates cell migration, establishing an early role for this signaling pathway. These results reveal a surprising departure from established mechanisms of axis formation: Huluwa-mediated prepatterning is dispensable, and ß-catenin and Nodal regulate morphogenesis.


Asunto(s)
Fundulidae , Morfogénesis , Proteína Nodal , beta Catenina , Animales , beta Catenina/metabolismo , Blástula/metabolismo , Tipificación del Cuerpo , Movimiento Celular , Núcleo Celular/metabolismo , Fundulidae/embriología , Fundulidae/metabolismo , Estratos Germinativos/metabolismo , Proteína Nodal/metabolismo , Transducción de Señal
8.
Nat Commun ; 15(1): 5055, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38871742

RESUMEN

The anterior-posterior axis of the mammalian embryo is laid down by the anterior visceral endoderm (AVE), an extraembryonic signaling center that is specified within the visceral endoderm. Current models posit that AVE differentiation is promoted globally by epiblast-derived Nodal signals, and spatially restricted by a BMP gradient established by the extraembryonic ectoderm. Here, we report spatially restricted AVE differentiation in bilayered embryo-like aggregates made from mouse embryonic stem cells that lack an extraembryonic ectoderm. Notably, clusters of AVE cells also form in pure visceral endoderm cultures upon activation of Nodal signaling, indicating that tissue-intrinsic factors can restrict AVE differentiation. We identify ß-catenin activity as a tissue-intrinsic factor that antagonizes AVE-inducing Nodal signals. Together, our results show how an AVE-like population can arise through interactions between epiblast and visceral endoderm alone. This mechanism may be a flexible solution for axis patterning in a wide range of embryo geometries, and provide robustness to axis patterning when coupled with signal gradients.


Asunto(s)
Tipificación del Cuerpo , Diferenciación Celular , Endodermo , Proteína Nodal , Transducción de Señal , beta Catenina , Animales , Endodermo/citología , Endodermo/metabolismo , Endodermo/embriología , beta Catenina/metabolismo , Ratones , Proteína Nodal/metabolismo , Proteína Nodal/genética , Estratos Germinativos/metabolismo , Estratos Germinativos/citología , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/citología , Regulación del Desarrollo de la Expresión Génica , Embrión de Mamíferos/citología
9.
PLoS Biol ; 22(6): e3002701, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38913712

RESUMEN

In the highly regulative embryo of the sea urchin Paracentrotus lividus, establishment of the dorsal-ventral (D/V) axis critically depends on the zygotic expression of the TGF-ß nodal in the ventral ectoderm. nodal expression is first induced ubiquitously in the 32-cell embryo and becomes progressively restricted to the presumptive ventral ectoderm by the early blastula stage. This early spatial restriction of nodal expression is independent of Lefty, and instead relies on the activity of Panda, a maternally expressed TGF-ß ligand related to Lefty and Inhibins, which is required maternally for D/V axis specification. However, the mechanism by which Panda restricts the early nodal expression has remained enigmatic and it is not known if Panda works like a BMP ligand by opposing Nodal and antagonizing Smad2/3 signaling, or if it works like Lefty by sequestering an essential component of the Nodal signaling pathway. In this study, we report that Panda functions as an antagonist of the TGF-ß type II receptor ACVRII (Activin receptor type II), which is the only type II receptor for Nodal signaling in the sea urchin and is also a type II receptor for BMP ligands. Inhibiting translation of acvrII mRNA disrupted D/V patterning across all 3 germ layers and caused acvrII morphants to develop with a typical Nodal loss-of-function phenotype. In contrast, embryos overexpressing acvrII displayed strong ectopic Smad1/5/8 signaling at blastula stages and developed as dorsalized larvae, a phenotype very similar to that caused by over activation of BMP signaling. Remarkably, embryos co-injected with acvrII mRNA and panda mRNA did not show ectopic Smad1/5/8 signaling and developed with a largely normal dorsal-ventral polarity. Furthermore, using an axis induction assay, we found that Panda blocks the ability of ACVRII to orient the D/V axis when overexpressed locally. Using co-immunoprecipitation, we showed that Panda physically interacts with ACVRII, as well as with the Nodal co-receptor Cripto, and with TBR3 (Betaglycan), which is a non-signaling receptor for Inhibins in mammals. At the molecular level, we have traced back the antagonistic activity of Panda to the presence of a single proline residue, conserved with all the Lefty factors, in the ACVRII binding motif of Panda, instead of a serine as in most of TGF-ß ligands. Conversion of this proline to a serine converted Panda from an antagonist that opposed Nodal signaling and promoted dorsalization to an agonist that promoted Nodal signaling and triggered ventralization when overexpressed. Finally, using phylogenomics, we analyzed the emergence of the agonist and antagonist form of Panda in the course of evolution. Our data are consistent with the idea that the presence of a serine at that position, like in most TGF-ß, was the ancestral condition and that the initial function of Panda was possibly in promoting and not in antagonizing Nodal signaling. These results highlight the existence of key functional and structural elements conserved between Panda and Lefty, allow to draw an intriguing parallel between sea urchin Panda and mammalian Inhibin α and raise the unexpected possibility that the original function of Panda may have been in activation of the Nodal pathway rather than in its inhibition.


Asunto(s)
Receptores de Activinas Tipo II , Tipificación del Cuerpo , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Proteína Nodal , Paracentrotus , Factor de Crecimiento Transformador beta , Animales , Factor de Crecimiento Transformador beta/metabolismo , Tipificación del Cuerpo/genética , Paracentrotus/embriología , Paracentrotus/metabolismo , Paracentrotus/genética , Receptores de Activinas Tipo II/metabolismo , Receptores de Activinas Tipo II/genética , Proteína Nodal/metabolismo , Proteína Nodal/genética , Embrión no Mamífero/metabolismo , Ligandos , Transducción de Señal
10.
Int J Biol Macromol ; 274(Pt 2): 133294, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38925188

RESUMEN

Congenital heart disease (CHD) is a type of major defect that occurs during embryonic development. Although significant advances have been made in the treatment of CHD, its etiology and molecular mechanism remain unclear. To identify the critical role of SUMOylation in cardiac development, we generated SENP3 knockout mice and showed that SENP3 knockout mice die on embryonic day 8.5 with an open neural tube and reversed left-right cardiac asymmetry. Moreover, SENP3 knockout promoted apoptosis and senescence of H9C2 cells. Further studies showed that Nodal, a critical gene that forms left-right asymmetry, is regulated by SENP3 and that SENP3 regulates cell apoptosis and senescence in a Nodal-dependent manner. Furthermore, Nodal was hyper-SUMOylated after SENP3 knockout, and SUMOylation of Nodal inhibited its ubiquitination and ubiquitin-proteasome degradation pathway. Nodal overexpression enhanced cell apoptosis and senescence; however, the mutation at the SUMOylation site of Nodal reversed its effect on the apoptosis and senescence of H9C2 cells. More importantly, the SENP3-Nodal axis regulates cell senescence by inducing cell autophagy. These results suggest that the SENP3-Nodal signaling axis regulates cardiac senescence-autophagy homeostasis, which in turn affects cardiac development and results in the occurrence of CHD.


Asunto(s)
Apoptosis , Cisteína Endopeptidasas , Proteína Nodal , Transducción de Señal , Sumoilación , Animales , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/genética , Ratones , Apoptosis/genética , Proteína Nodal/metabolismo , Proteína Nodal/genética , Corazón/embriología , Ratones Noqueados , Autofagia/genética , Senescencia Celular/genética , Línea Celular
11.
J Cell Physiol ; 239(6): e31268, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38577903

RESUMEN

Several members of the transforming growth factor beta (TGF-ß) superfamily regulate the proliferation, differentiation, and function of bone-forming osteoblasts and bone-resorbing osteoclasts. However, it is still unknown whether Nodal, a member of the TGF-ß superfamily, serves a function in bone cells. In this study, we found that Nodal did not have any function in osteoblasts but instead negatively regulated osteoclast differentiation. Nodal inhibited RANKL-induced osteoclast differentiation by downregulating the expression of pro-osteoclastogenic genes, including c-fos, Nfatc1, and Blimp1, and upregulating the expression of antiosteoclastogenic genes, including Bcl6 and Irf8. Nodal activated STAT1 in osteoclast precursor cells, and STAT1 downregulation significantly reduced the inhibitory effect of Nodal on osteoclast differentiation. These findings indicate that Nodal activates STAT1 to downregulate or upregulate the expression of pro-osteoclastogenic or antiosteoclastogenic genes, respectively, leading to the inhibition of osteoclast differentiation. Moreover, the inhibitory effect of Nodal on osteoclast differentiation contributed to the reduction of RANKL-induced bone loss in vivo.


Asunto(s)
Diferenciación Celular , Proteína Nodal , Osteoclastos , Factor de Transcripción STAT1 , Animales , Ratones , Resorción Ósea/metabolismo , Resorción Ósea/genética , Resorción Ósea/patología , Factores Reguladores del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Factores de Transcripción NFATC/metabolismo , Factores de Transcripción NFATC/genética , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogénesis/genética , Fosforilación , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Ligando RANK/metabolismo , Transducción de Señal , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT1/genética , Masculino , Ratones Endogámicos ICR , Proteína Nodal/genética , Proteína Nodal/metabolismo , Proteína Nodal/farmacología
12.
Methods Mol Biol ; 2803: 13-33, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38676882

RESUMEN

The adept and systematic differentiation of embryonic stem cells (ESCs) and human-induced pluripotent stem cells (hiPSCs) to diverse lineage-prone cell types involves crucial step-by-step process that mimics the vital strategic commitment phase that is usually observed during the process of embryo development. The development of precise tissue-specific cell types from these stem cells indeed plays an important role in the advancement of imminent stem cell-based therapeutic strategies. Therefore, the usage of hiPSC-derived cell types for subsequent cardiovascular disease modeling, drug screening, and therapeutic drug development undeniably entails an in-depth understanding of each and every step to proficiently stimulate these stem cells into desired cardiomyogenic lineage. Thus, to accomplish this definitive and decisive fate, it is essential to efficiently induce the mesoderm or pre-cardiac mesoderm, succeeded by the division of cells into cardiovascular and ultimately ensuing with the cardiomyogenic lineage outcome. This usually commences from the earliest phases of pluripotent cell induction. In this chapter, we discuss our robust and reproducible step-wise protocol that will describe the subtype controlled, precise lineage targeted standardization of activin/nodal, and BMP signaling molecules/cytokines, for the efficient differentiation of ventricular cardiomyocytes from hiPSCs via the embryoid body method. In addition, we also describe techniques to dissociate hiPSCs, hiPSC-derived early cardiomyocytes for mesoderm and pre-cardiac mesoderm assessment, and hiPSC-derived cardiomyocytes for early and mature markers assessment.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Humanos , Activinas/farmacología , Activinas/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Técnicas de Cultivo de Célula/métodos , Linaje de la Célula , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteína Nodal/metabolismo , Transducción de Señal
13.
Development ; 151(4)2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38372390

RESUMEN

Embryogenesis results from the coordinated activities of different signaling pathways controlling cell fate specification and morphogenesis. In vertebrate gastrulation, both Nodal and BMP signaling play key roles in germ layer specification and morphogenesis, yet their interplay to coordinate embryo patterning with morphogenesis is still insufficiently understood. Here, we took a reductionist approach using zebrafish embryonic explants to study the coordination of Nodal and BMP signaling for embryo patterning and morphogenesis. We show that Nodal signaling triggers explant elongation by inducing mesendodermal progenitors but also suppressing BMP signaling activity at the site of mesendoderm induction. Consistent with this, ectopic BMP signaling in the mesendoderm blocks cell alignment and oriented mesendoderm intercalations, key processes during explant elongation. Translating these ex vivo observations to the intact embryo showed that, similar to explants, Nodal signaling suppresses the effect of BMP signaling on cell intercalations in the dorsal domain, thus allowing robust embryonic axis elongation. These findings suggest a dual function of Nodal signaling in embryonic axis elongation by both inducing mesendoderm and suppressing BMP effects in the dorsal portion of the mesendoderm.


Asunto(s)
Tipificación del Cuerpo , Pez Cebra , Animales , Tipificación del Cuerpo/genética , Proteína Nodal/genética , Proteína Nodal/metabolismo , Morfogénesis/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Regulación del Desarrollo de la Expresión Génica
14.
Front Immunol ; 14: 1276979, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38022561

RESUMEN

Pregnancy success is dependent on the establishment of maternal tolerance during the preimplantation period. The immunosuppressive function of regulatory T cells is critical to limit inflammation arising from implantation of the semi-allogeneic blastocyst. Insufficient maternal immune adaptations to pregnancy have been frequently associated with cases of female infertility and recurrent implantation failure. The role of Nodal, a secreted morphogen of the TGFß superfamily, was recently implicated during murine pregnancy as its conditional deletion (NodalΔ/Δ) in the female reproductive tract resulted in severe subfertility. Here, it was determined that despite normal preimplantation processes and healthy, viable embryos, NodalΔ/Δ females had a 50% implantation failure rate compared to NodalloxP/loxP controls. Prior to implantation, the expression of inflammatory cytokines MCP-1, G-CSF, IFN-γ and IL-10 was dysregulated in the NodalΔ/Δ uterus. Further analysis of the preimplantation leukocyte populations in NodalΔ/Δ uteri showed an overabundance of infiltrating, pro-inflammatory CD11bhigh Ly6C+ macrophages coupled with the absence of CD4+ FOXP3+ regulatory T cells. Therefore, it is proposed that uterine Nodal expression during the preimplantation period has a novel role in the establishment of maternal immunotolerance, and its dysregulation should be considered as a potential contributor to cases of female infertility and recurrent implantation failure.


Asunto(s)
Infertilidad Femenina , Proteína Nodal , Linfocitos T Reguladores , Animales , Femenino , Ratones , Embarazo , Implantación del Embrión , Factores de Transcripción Forkhead , Infertilidad Femenina/genética , Útero , Proteína Nodal/genética
15.
Dev Cell ; 58(16): 1447-1461.e6, 2023 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-37413993

RESUMEN

Left-dominant [Ca2+]i elevation on the left margin of the ventral node furnishes the initial laterality of mouse embryos. It depends on extracellular leftward fluid flow (nodal flow), fibroblast growth factor receptor (FGFR)/sonic hedgehog (Shh) signaling, and the PKD1L1 polycystin subunit, of which interrelationship is still elusive. Here, we show that leftward nodal flow directs PKD1L1-containing fibrous strands and facilitates Nodal-mediated [Ca2+]i elevation on the left margin. We generate KikGR-PKD1L1 knockin mice in order to monitor protein dynamics with a photoconvertible fluorescence protein tag. By imaging those embryos, we have identified fragile meshwork being gradually transferred leftward involving pleiomorphic extracellular events. A portion of the meshwork finally bridges over the left nodal crown cells in an FGFR/Shh-dependent manner. As PKD1L1 N-term is predominantly associated with Nodal on the left margin and that PKD1L1/PKD2 overexpression significantly augments cellular Nodal sensitivity, we propose that leftward transfer of polycystin-containing fibrous strands determines left-right asymmetry in developing embryos.


Asunto(s)
Proteínas Hedgehog , Canales Catiónicos TRPP , Ratones , Animales , Proteínas Hedgehog/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo , Cilios/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Tipificación del Cuerpo , Regulación del Desarrollo de la Expresión Génica , Proteína Nodal/metabolismo
16.
Glia ; 71(9): 2096-2116, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37208933

RESUMEN

Our prior work examining endogenous repair after spinal cord injury (SCI) in mice revealed that large numbers of new oligodendrocytes (OLs) are generated in the injured spinal cord, with peak oligodendrogenesis between 4 and 7 weeks post-injury (wpi). We also detected new myelin formation over 2 months post-injury (mpi). Our current work significantly extends these results, including quantification of new myelin through 6 mpi and concomitant examination of indices of demyelination. We also examined electrophysiological changes during peak oligogenesis and a potential mechanism driving OL progenitor cell (OPC) contact with axons. Results reveal peak in remyelination occurs during the 3rd mpi, and that myelin generation continues for at least 6 mpi. Further, motor evoked potentials significantly increased during peak remyelination, suggesting enhanced axon potential conduction. Interestingly, two indices of demyelination, nodal protein spreading and Nav1.2 upregulation, were also present chronically after SCI. Nav1.2 was expressed through 10 wpi and nodal protein disorganization was detectable throughout 6 mpi suggesting chronic demyelination, which was confirmed with EM. Thus, demyelination may continue chronically, which could trigger the long-term remyelination response. To examine a potential mechanism that may initiate post-injury myelination, we show that OPC processes contact glutamatergic axons in the injured spinal cord in an activity-dependent manner. Notably, these OPC/axon contacts were increased 2-fold when axons were activated chemogenetically, revealing a potential therapeutic target to enhance post-SCI myelin repair. Collectively, results show the surprisingly dynamic nature of the injured spinal cord over time and that the tissue may be amenable to treatments targeting chronic demyelination.


Asunto(s)
Enfermedades Desmielinizantes , Traumatismos de la Médula Espinal , Ratones , Animales , Vaina de Mielina/metabolismo , Proteína Nodal/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Axones/fisiología , Oligodendroglía/metabolismo , Médula Espinal , Enfermedades Desmielinizantes/metabolismo
17.
Clin Neurol Neurosurg ; 223: 107521, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36401951

RESUMEN

OBJECTIVE: Antibodies against nodal-paranodal junction proteins have been detected in some patients with chronic inflammatory demyelinating polyradiculoneuropathy (CIDP), which is a crucial step to define the most effective treatment strategies. In this paper, we tested the positive rates of these antibodies in CIDP and characterized the clinical and electrophysiological features of the antibodies-positive patients. METHODS: We prospectively recruited 47 patients with CIDP. We detected IgG antibodies against human neurofascin-155 (NF155), neurofascin-186 (NF186), contactin-1 (CNTN1), contactin-2 (CNTN2) and contactin-associated protein-1 (Caspr1), and identified the IgG isotype with cell-based assay (CBA). We collected the demographic, clinical, laboratory, and electrophysiological information of the patients that were seropositive. RESULTS: Five patients (10.6 %) had IgG against NF155, 3 (6.4 %) against Caspr1, 2 (4.3 %) against NF186 and 1 (2.1 %) against CNTN1. All the 11 antibody-positive patients (8 males and 3 females) presented with typical clinical features. Five of them needed assistance in walking, 5 had cranial nerve impairments and 3 had autonomic disturbances. The age at onset of the patients that were anti-NF155-positive was younger (19.60 ± 9.02 years vs. 55.33 ± 11.93 years, P = 0.003) than those that were anti-Caspr1-positive. No significant difference in the functional status was observed between these two groups. The action potentials of 11/79 (13.9 %) motor nerves and 62/93 (66.7 %) sensory nerves exhibited no response. Moreover, 16/68 (23.5 %) nerves presented conduction block and 13/68 (19.1 %) nerves presented temporal dispersion. Distal motor latency (DML) of ulnar nerve and tibial nerve tended to be longer (p = 0.008 and p = 0.006, respectively) in anti-NF155-positive patients than that in anti-Caspr1-positive patients. Of the 11 patients that were antibody-positive patients, corticosteroids were effective in 3/7 (42.9 %), intravenous immunoglobins (IVIG) were effective in 1/7 (14.3 %), and rituximab was effective in 6/8 (75.0 %). CONCLUSIONS: Our findings validate the previous observation on the clinico-serological correlation between CIDP and antibodies against nodal-paranodal proteins. Of note, the damage on nerves is more severe in anti-NF155-positive patients than that in anti-Caspr1-positive patients during electrophysiological diagnosis.


Asunto(s)
Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante , Adolescente , Adulto , Niño , Femenino , Humanos , Masculino , Adulto Joven , Contactinas , Nervios Craneales , Inmunoglobulina G , Proteína Nodal , Persona de Mediana Edad , Anciano
18.
Nat Commun ; 13(1): 6101, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36243734

RESUMEN

The hindered diffusion model postulates that the movement of a signaling molecule through an embryo is affected by tissue geometry and binding-mediated hindrance, but these effects have not been directly demonstrated in vivo. Here, we visualize extracellular movement and binding of individual molecules of the activator-inhibitor signaling pair Nodal and Lefty in live developing zebrafish embryos using reflected light-sheet microscopy. We observe that diffusion coefficients of molecules are high in extracellular cavities, whereas mobility is reduced and bound fractions are high within cell-cell interfaces. Counterintuitively, molecules nevertheless accumulate in cavities, which we attribute to the geometry of the extracellular space by agent-based simulations. We further find that Nodal has a larger bound fraction than Lefty and shows a binding time of tens of seconds. Together, our measurements and simulations provide direct support for the hindered diffusion model and yield insights into the nanometer-to-micrometer-scale mechanisms that lead to macroscopic signal dispersal.


Asunto(s)
Proteína Nodal , Pez Cebra , Animales , Difusión , Regulación del Desarrollo de la Expresión Génica , Factores de Determinación Derecha-Izquierda/genética , Proteína Nodal/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/genética
19.
Elife ; 112022 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-36149406

RESUMEN

During vertebrate embryogenesis, the germ layers are patterned by secreted Nodal signals. In the classical model, Nodals elicit signaling by binding to a complex comprising Type I/II Activin receptors (Acvr) and the co-receptor Tdgf1. However, it is currently unclear whether receptor binding can also affect the distribution of Nodals themselves through the embryo, and it is unknown which of the putative Acvr paralogs mediate Nodal signaling in zebrafish. Here, we characterize three Type I (Acvr1) and four Type II (Acvr2) homologs and show that - except for Acvr1c - all receptor-encoding transcripts are maternally deposited and present during zebrafish embryogenesis. We generated mutants and used them together with combinatorial morpholino knockdown and CRISPR F0 knockout (KO) approaches to assess compound loss-of-function phenotypes. We discovered that the Acvr2 homologs function partly redundantly and partially independently of Nodal to pattern the early zebrafish embryo, whereas the Type I receptors Acvr1b-a and Acvr1b-b redundantly act as major mediators of Nodal signaling. By combining quantitative analyses with expression manipulations, we found that feedback-regulated Type I receptors and co-receptors can directly influence the diffusion and distribution of Nodals, providing a mechanism for the spatial restriction of Nodal signaling during germ layer patterning.


Building a body is complicated. Cells must organise themselves head-to-tail, belly-to-back, and inside-to-outside. They do this by laying down a chemical map, which is made up of gradients of molecular signals, high in some places and lower in others. The amount of signal each cell receives helps to decide which part of the body it will become. One of the essential signals in developing vertebrates is Nodal. It helps cells to tell inside from outside and left from right. Cells detect Nodal using an activin receptor and co-receptor complex, which catch hold of passing Nodal proteins and transmit developmental signals into cells. An important model to study Nodal signals is the zebrafish embryo, but the identity of the activin receptors and their exact role in this organism has been unclear. To find out more, Preiß, Kögler, Mörsdorf et al. studied the activin receptors Acvr1 and Acvr2 in zebrafish embryos. The experiments revealed that two putative Acvr1 and four Acvr2 receptors were present during early development. To better understand their roles, Preiß et al. eliminated them one at a time, and in combination. Losing single activin receptors had no effect. But losing both Acvr1 receptors together stopped Nodal signalling and changed the distribution of the Nodal gradient. Loss of all Acvr2 receptors also caused developmental problems, but they were partly independent of Nodal. This suggests that Acvr1s seem to be able to transmit signals and to shape the Nodal gradient, and that Acvr2s might have another, so far unknown, role. Nodal signals guide the development of all vertebrates. Understanding how they work in a model species like zebrafish could shed light on their role in other species, including humans. A clearer picture could help to uncover what happens at a molecular level when development goes wrong.


Asunto(s)
Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/genética , Pez Cebra/metabolismo , Retroalimentación , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Proteína Nodal/genética , Proteína Nodal/metabolismo , Tipificación del Cuerpo/genética , Regulación del Desarrollo de la Expresión Génica
20.
Science ; 377(6613): eabl3921, 2022 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-36137018

RESUMEN

The vertebrate intestine forms by asymmetric gut rotation and elongation, and errors cause lethal obstructions in human infants. Rotation begins with tissue deformation of the dorsal mesentery, which is dependent on left-sided expression of the Paired-like transcription factor Pitx2. The conserved morphogen Nodal induces asymmetric Pitx2 to govern embryonic laterality, but organ-level regulation of Pitx2 during gut asymmetry remains unknown. We found Nodal to be dispensable for Pitx2 expression during mesentery deformation. Intestinal rotation instead required a mechanosensitive latent transforming growth factor-ß (TGFß), tuning a second wave of Pitx2 that induced reciprocal tissue stiffness in the left mesentery as mechanical feedback with the right side. This signaling regulator, an accelerator (right) and brake (left), combines biochemical and biomechanical inputs to break gut morphological symmetry and direct intestinal rotation.


Asunto(s)
Gastrulación , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio , Intestinos , Mecanotransducción Celular , Proteína Nodal , Factores de Transcripción , Factor de Crecimiento Transformador beta , Animales , Embrión de Pollo , Gastrulación/genética , Gastrulación/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/farmacología , Intestinos/embriología , Mecanotransducción Celular/genética , Mecanotransducción Celular/fisiología , Ratones , Proteína Nodal/genética , Factores de Transcripción/genética , Factores de Transcripción/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Proteína del Homeodomínio PITX2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA