Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(42)2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34593624

RESUMEN

The coronaviruses responsible for severe acute respiratory syndrome (SARS-CoV), COVID-19 (SARS-CoV-2), Middle East respiratory syndrome-CoV, and other coronavirus infections express a nucleocapsid protein (N) that is essential for viral replication, transcription, and virion assembly. Phosphorylation of N from SARS-CoV by glycogen synthase kinase 3 (GSK-3) is required for its function and inhibition of GSK-3 with lithium impairs N phosphorylation, viral transcription, and replication. Here we report that the SARS-CoV-2 N protein contains GSK-3 consensus sequences and that this motif is conserved in diverse coronaviruses, raising the possibility that SARS-CoV-2 may be sensitive to GSK-3 inhibitors, including lithium. We conducted a retrospective analysis of lithium use in patients from three major health systems who were PCR-tested for SARS-CoV-2. We found that patients taking lithium have a significantly reduced risk of COVID-19 (odds ratio = 0.51 [0.35-0.74], P = 0.005). We also show that the SARS-CoV-2 N protein is phosphorylated by GSK-3. Knockout of GSK3A and GSK3B demonstrates that GSK-3 is essential for N phosphorylation. Alternative GSK-3 inhibitors block N phosphorylation and impair replication in SARS-CoV-2 infected lung epithelial cells in a cell-type-dependent manner. Targeting GSK-3 may therefore provide an approach to treat COVID-19 and future coronavirus outbreaks.


Asunto(s)
COVID-19/prevención & control , Proteínas de la Nucleocápside de Coronavirus/metabolismo , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Compuestos de Litio/uso terapéutico , Adulto , Anciano , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Células HEK293 , Humanos , Compuestos de Litio/farmacología , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Estudios Retrospectivos
2.
Development ; 146(8)2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30910828

RESUMEN

In most species, early germline development occurs in the absence of transcription with germline determinants subject to complex translational and post-translational regulations. Here, we report for the first time that early germline development is influenced by dynamic regulation of the proteasome system, previously thought to be ubiquitously expressed and to serve 'housekeeping' roles in controlling protein homeostasis. We show that proteasomes are present in a gradient with the highest levels in the animal hemisphere and extending into the vegetal hemisphere of Xenopus oocytes. This distribution changes dramatically during the oocyte-to-embryo transition, with proteasomes becoming enriched in and restricted to the animal hemisphere and therefore separated from vegetally localized germline determinants. We identify Dead-end1 (Dnd1), a master regulator of vertebrate germline development, as a novel substrate of the ubiquitin-independent proteasomes. In the oocyte, ubiquitin-independent proteasomal degradation acts together with translational repression to prevent premature accumulation of Dnd1 protein. In the embryo, artificially increasing ubiquitin-independent proteasomal degradation in the vegetal pole interferes with germline development. Our work thus reveals novel inhibitory functions and spatial regulation of the ubiquitin-independent proteasome during vertebrate germline development.


Asunto(s)
Células Germinativas/metabolismo , Ubiquitina/metabolismo , Animales , Citoplasma/metabolismo , Células Germinativas/citología , Oocitos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas de Unión al ARN/metabolismo , Ubiquitina/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis
3.
Dev Biol ; 462(1): 20-35, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32119833

RESUMEN

As development proceeds, inductive cues are interpreted by competent tissues in a spatially and temporally restricted manner. While key inductive signaling pathways within competent cells are well-described at a molecular level, the mechanisms by which tissues lose responsiveness to inductive signals are not well understood. Localized activation of Wnt signaling before zygotic gene activation in Xenopus laevis leads to dorsal development, but competence to induce dorsal genes in response to Wnts is lost by the late blastula stage. We hypothesize that loss of competence is mediated by changes in histone modifications leading to a loss of chromatin accessibility at the promoters of Wnt target genes. We use ATAC-seq to evaluate genome-wide changes in chromatin accessibility across several developmental stages. Based on overlap with p300 binding, we identify thousands of putative cis-regulatory elements at the gastrula stage, including sites that lose accessibility by the end of gastrulation and are enriched for pluripotency factor binding motifs. Dorsal Wnt target gene promoters are not accessible after the loss of competence in the early gastrula while genes involved in mesoderm and neural crest development maintain accessibility at their promoters. Inhibition of histone deacetylases increases acetylation at the promoters of dorsal Wnt target genes and extends competence for dorsal gene induction by Wnt signaling. Histone deacetylase inhibition, however, is not sufficient to extend competence for mesoderm or neural crest induction. These data suggest that chromatin state regulates the loss of competence to inductive signals in a context-dependent manner.


Asunto(s)
Cromatina/metabolismo , Inducción Embrionaria/genética , Histonas/metabolismo , Acetilación , Animales , Blástula/metabolismo , Cromatina/genética , Gástrula/metabolismo , Gastrulación/genética , Regulación del Desarrollo de la Expresión Génica/genética , Histonas/genética , Mesodermo/metabolismo , Cresta Neural/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/fisiología , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Xenopus laevis/metabolismo
4.
Anal Chem ; 91(14): 8891-8899, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31194517

RESUMEN

Techniques that allow single cell analysis are gaining widespread attention, and most of these studies utilize genomics-based approaches. While nanofluidic technologies have enabled mass spectrometric analysis of single cells, these measurements have been limited to metabolomics and lipidomic studies. Single cell proteomics has the potential to improve our understanding of intercellular heterogeneity. However, this approach has faced challenges including limited sample availability, as well as a requirement of highly sensitive methods for sample collection, cleanup, and detection. We present a technique to overcome these limitations by combining a micropipette (pulled glass capillary) based sample collection strategy with offline sample preparation and nanoLC-MS/MS to analyze proteins through a bottom-up proteomic strategy. This study explores two types of proteomics data acquisition strategies namely data-dependent (DDA) and data-independent acquisition (DIA). Results from the study indicate DIA to be more sensitive enabling analysis of >1600 proteins from ∼130 µm Xenopus laevis embryonic cells containing <6 nL of cytoplasm. The method was found to be robust in obtaining reproducible protein quantifications from single cells spanning the 1-128-cell stages of development. Furthermore, we used micropipette sampling to study intercellular heterogeneity within cells in a single embryo and investigated embryonic asymmetry along both animal-vegetal and dorsal-ventral axes during early stages of development. Investigation of the animal-vegetal axis led to discovery of various asymmetrically distributed proteins along the animal-vegetal axis. We have further compared the hits found from our proteomic data sets with other studies and validated a few hits using an orthogonal imaging technique. This study forms the first report of vegetal enrichment of the germ plasm associated protein DDX4/VASA in Xenopus embyos. Overall, the method and data presented here holds promise to enable important leads in developmental biology.


Asunto(s)
Embrión no Mamífero/citología , Proteómica/métodos , Análisis de la Célula Individual/métodos , Proteínas de Xenopus/análisis , Xenopus laevis/embriología , Animales , Embrión no Mamífero/química , Espectrometría de Masas en Tándem/métodos
5.
J Biol Chem ; 292(44): 18240-18255, 2017 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-28916722

RESUMEN

Glycogen synthase kinase-3 (GSK-3) is a constitutively active, ubiquitously expressed protein kinase that regulates multiple signaling pathways. In vitro kinase assays and genetic and pharmacological manipulations of GSK-3 have identified more than 100 putative GSK-3 substrates in diverse cell types. Many more have been predicted on the basis of a recurrent GSK-3 consensus motif ((pS/pT)XXX(S/T)), but this prediction has not been tested by analyzing the GSK-3 phosphoproteome. Using stable isotope labeling of amino acids in culture (SILAC) and MS techniques to analyze the repertoire of GSK-3-dependent phosphorylation in mouse embryonic stem cells (ESCs), we found that ∼2.4% of (pS/pT)XXX(S/T) sites are phosphorylated in a GSK-3-dependent manner. A comparison of WT and Gsk3a;Gsk3b knock-out (Gsk3 DKO) ESCs revealed prominent GSK-3-dependent phosphorylation of multiple splicing factors and regulators of RNA biosynthesis as well as proteins that regulate transcription, translation, and cell division. Gsk3 DKO reduced phosphorylation of the splicing factors RBM8A, SRSF9, and PSF as well as the nucleolar proteins NPM1 and PHF6, and recombinant GSK-3ß phosphorylated these proteins in vitro RNA-Seq of WT and Gsk3 DKO ESCs identified ∼190 genes that are alternatively spliced in a GSK-3-dependent manner, supporting a broad role for GSK-3 in regulating alternative splicing. The MS data also identified posttranscriptional regulation of protein abundance by GSK-3, with ∼47 proteins (1.4%) whose levels increased and ∼78 (2.4%) whose levels decreased in the absence of GSK-3. This study provides the first unbiased analysis of the GSK-3 phosphoproteome and strong evidence that GSK-3 broadly regulates alternative splicing.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Nucleares/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas de Unión al ARN/metabolismo , Factores de Empalme Serina-Arginina/metabolismo , Empalme Alternativo , Animales , Isótopos de Carbono , Línea Celular , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/enzimología , Células Madre Embrionarias/metabolismo , Técnicas de Inactivación de Genes , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta/genética , Proteínas de Homeodominio/química , Ratones , Isótopos de Nitrógeno , Proteínas Nucleares/química , Nucleofosmina , Mapeo Peptídico , Fosforilación , Estabilidad Proteica , Proteómica/métodos , Proteínas de Unión al ARN/química , Proteínas Recombinantes/metabolismo , Proteínas Represoras , Factores de Empalme Serina-Arginina/química , Especificidad por Sustrato
6.
Adv Exp Med Biol ; 953: 441-487, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27975277

RESUMEN

Following fertilization, vertebrate embryos delay large-scale activation of the zygotic genome from several hours in fish and amphibians to several days in mammals. Externally developing embryos also undergo synchronous and extraordinarily rapid cell divisions that are accelerated by promiscuous licensing of DNA replication origins, absence of gap phases and cell cycle checkpoints, and preloading of the egg with maternal RNAs and proteins needed to drive early development. After a species-specific number of cell divisions, the cell cycle slows and becomes asynchronous, gap phases appear, checkpoint functions are acquired, and large-scale zygotic gene activation begins. These events, along with clearance of maternal RNAs and proteins, define the maternal to zygotic transition and are coordinated at a developmental milestone termed the midblastula transition (MBT). Despite the relative quiescence of the zygotic genome in vertebrate embryos, genes required for clearance of maternal RNAs and for the initial steps in mesoderm induction are robustly transcribed before MBT. The coordination and timing of the MBT depends on a mechanism that senses the ratio of nuclear to cytoplasmic content as well as mechanisms that are independent of the nuclear-cytoplasm ratio. Changes in chromatin architecture anticipate zygotic gene activation, and maternal transcription factors identified as regulators of pluripotency play critical roles in kick-starting the transition from the proliferative, pluripotent state of the early embryo to the more lineage-committed phase of development after the MBT. This chapter describes the regulation of the cell cycle and the activation of zygotic gene expression before and after the MBT in vertebrate embryos.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , Desarrollo Embrionario/genética , Activación Transcripcional/genética , Cigoto/crecimiento & desarrollo , Animales , Ciclo Celular/genética , División Celular/genética , Femenino , Regulación del Desarrollo de la Expresión Génica
7.
Stem Cells ; 33(1): 278-88, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25284014

RESUMEN

Hematopoiesis is maintained throughout life by self-renewing hematopoietic stem cells (HSCs) that differentiate to produce both myeloid and lymphoid cells. The NR4A family of orphan nuclear receptors, which regulates cell fate in many tissues, appears to play a key role in HSC proliferation and differentiation. Using a NR4A1(GFP) BAC transgenic reporter mouse we have investigated NR4A1 expression and its regulation in early hematopoiesis. We show that NR4A1 is most highly expressed in a subset of Lin(-) Sca-1(+) c-Kit(+) CD48(-) CD150(+) long-term (LT) HSCs, and its expression is tightly associated with HSC quiescence. We also show that NR4A1 expression in HSCs is induced by PGE2, a known enhancer of stem cell engraftment potential. Finally, we find that both NR4A1(GFP+) and NR4A1(GFP-) HSCs successfully engraft primary and secondary irradiated hosts; however, NR4A1(GFP+) HSCs are distinctly myeloid-biased. These results show that NR4A1 expression identifies a highly quiescent and distinct population of myeloid-biased LT-HSCs.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Ratones , Ratones Endogámicos C57BL
8.
Dev Dyn ; 242(2): 108-21, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23184530

RESUMEN

BACKGROUND: Histone deacetylases (HDACs) regulate multiple developmental processes and cellular functions. However, their roles in blood development have not been determined, and in Xenopus laevis a specific function for HDACs has yet to be identified. Here, we employed the class I selective HDAC inhibitor, valproic acid (VPA), to show that HDAC activity is required for primitive hematopoiesis. RESULTS: VPA treatment during gastrulation resulted in a complete absence of red blood cells (RBCs) in Xenopus tadpoles, but did not affect development of other mesodermal tissues, including myeloid and endothelial lineages. These effects of VPA were mimicked by Trichostatin A (TSA), a well-established pan-HDAC inhibitor, but not by valpromide, which is structurally similar to VPA but does not inhibit HDACs. VPA also caused a marked, dose-dependent loss of primitive erythroid progenitors in mouse yolk sac explants at clinically relevant concentrations. In addition, VPA treatment inhibited erythropoietic development downstream of bmp4 and gata1 in Xenopus ectodermal explants. CONCLUSIONS: These findings suggest an important role for class I HDACs in primitive hematopoiesis. Our work also demonstrates that specific developmental defects associated with exposure to VPA, a significant teratogen in humans, arise through inhibition of class I HDACs.


Asunto(s)
Gástrula/efectos de los fármacos , Hematopoyesis/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Xenopus laevis/embriología , Animales , Cartilla de ADN/genética , Células Precursoras Eritroides/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Immunoblotting , Hibridación in Situ , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Ácido Valproico/farmacología , Saco Vitelino/citología
9.
J Clin Invest ; 134(12)2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38713535

RESUMEN

Splicing factor mutations are common in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML), but how they alter cellular functions is unclear. We show that the pathogenic SRSF2P95H/+ mutation disrupts the splicing of mitochondrial mRNAs, impairs mitochondrial complex I function, and robustly increases mitophagy. We also identified a mitochondrial surveillance mechanism by which mitochondrial dysfunction modifies splicing of the mitophagy activator PINK1 to remove a poison intron, increasing the stability and abundance of PINK1 mRNA and protein. SRSF2P95H-induced mitochondrial dysfunction increased PINK1 expression through this mechanism, which is essential for survival of SRSF2P95H/+ cells. Inhibition of splicing with a glycogen synthase kinase 3 inhibitor promoted retention of the poison intron, impairing mitophagy and activating apoptosis in SRSF2P95H/+ cells. These data reveal a homeostatic mechanism for sensing mitochondrial stress through PINK1 splicing and identify increased mitophagy as a disease marker and a therapeutic vulnerability in SRSF2P95H mutant MDS and AML.


Asunto(s)
Leucemia Mieloide Aguda , Mitocondrias , Mitofagia , Proteínas Quinasas , Factores de Empalme Serina-Arginina , Animales , Humanos , Ratones , Sustitución de Aminoácidos , Línea Celular Tumoral , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patología , Neoplasias Hematológicas/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Mitofagia/genética , Mutación Missense , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Síndromes Mielodisplásicos/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Empalme del ARN , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo
10.
bioRxiv ; 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38712254

RESUMEN

Splicing factor mutations are common in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML), but how they alter cellular functions is unclear. We show that the pathogenic SRSF2P95H/+ mutation disrupts the splicing of mitochondrial mRNAs, impairs mitochondrial complex I function, and robustly increases mitophagy. We also identified a mitochondrial surveillance mechanism by which mitochondrial dysfunction modifies splicing of the mitophagy activator PINK1 to remove a poison intron, increasing the stability and abundance of PINK1 mRNA and protein. SRSF2P95H-induced mitochondrial dysfunction increased PINK1 expression through this mechanism, which is essential for survival of SRSF2P95H/+ cells. Inhibition of splicing with a glycogen synthase kinase 3 inhibitor promoted retention of the poison intron, impairing mitophagy and activating apoptosis in SRSF2P95H/+ cells. These data reveal a homeostatic mechanism for sensing mitochondrial stress through PINK1 splicing and identify increased mitophagy as a disease marker and a therapeutic vulnerability in SRSF2P95H mutant MDS and AML.

11.
J Biol Chem ; 287(6): 3823-32, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22184111

RESUMEN

Glycogen synthase kinase-3 (GSK-3) is essential for many signaling pathways and cellular processes. As Adenomatous Polyposis Coli (APC) functions in many of the same processes, we investigated a role for APC in the regulation of GSK-3-dependent signaling. We find that APC directly enhances GSK-3 activity. Furthermore, knockdown of APC mimics inhibition of GSK-3 by reducing phosphorylation of glycogen synthase and by activating mTOR, revealing novel roles for APC in the regulation of these enzymes. Wnt signaling inhibits GSK-3 through an unknown mechanism, and this results in both stabilization of ß-catenin and activation of mTOR. We therefore hypothesized that Wnts may regulate GSK-3 by disrupting the interaction between APC and the Axin-GSK-3 complex. We find that Wnts rapidly induce APC dissociation from Axin, correlating with ß-catenin stabilization. Furthermore, Axin interaction with the Wnt co-receptor LRP6 causes APC dissociation from Axin. We propose that APC regulates multiple signaling pathways by enhancing GSK-3 activity, and that Wnts induce APC dissociation from Axin to reduce GSK-3 activity and activate downstream signaling. APC regulation of GSK-3 also provides a novel mechanism for Wnt regulation of multiple downstream effectors, including ß-catenin and mTOR.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/fisiología , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína Axina/genética , Proteína Axina/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Células HEK293 , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Serina-Treonina Quinasas TOR/genética , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
12.
J Cell Sci ; 124(Pt 13): 2267-76, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21652627

RESUMEN

Valproic acid (VPA) is the most highly prescribed epilepsy treatment worldwide and is also used to prevent bipolar disorder and migraine. Surprisingly, very little is known about its mechanisms of cellular uptake. Here, we employ a range of cellular, molecular and genetic approaches to characterize VPA uptake using a simple biomedical model, Dictyostelium discoideum. We show that VPA is taken up against an electrochemical gradient in a dose-dependent manner. Transport is protein-mediated, dependent on pH and the proton gradient and shows strong substrate structure specificity. Using a genetic screen, we identified a protein homologous to a mammalian solute carrier family 4 (SLC4) bicarbonate transporter that we show is involved in VPA uptake. Pharmacological and genetic ablation of this protein reduces the uptake of VPA and partially protects against VPA-dependent developmental effects, and extracellular bicarbonate competes for VPA uptake in Dictyostelium. We further show that this uptake mechanism is likely to be conserved in both zebrafish (Danio rerio) and Xenopus laevis model systems. These results implicate, for the first time, an uptake mechanism for VPA through SLC4-catalysed activity.


Asunto(s)
Dictyostelium/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Ácido Valproico/metabolismo , Animales , Transporte Biológico Activo/fisiología , Células Cultivadas , Dictyostelium/efectos de los fármacos , Concentración de Iones de Hidrógeno , Proteínas de Transporte de Membrana/genética , Especificidad por Sustrato , Ácido Valproico/farmacología , Xenopus laevis , Pez Cebra
13.
Development ; 137(9): 1531-41, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20356957

RESUMEN

In vertebrates, canonical Wnt signaling controls posterior neural cell lineage specification. Although Wnt signaling to the neural plate is sufficient for posterior identity, the source and timing of this activity remain uncertain. Furthermore, crucial molecular targets of this activity have not been defined. Here, we identify the endogenous Wnt activity and its role in controlling an essential downstream transcription factor, Meis3. Wnt3a is expressed in a specialized mesodermal domain, the paraxial dorsolateral mesoderm, which signals to overlying neuroectoderm. Loss of zygotic Wnt3a in this region does not alter mesoderm cell fates, but blocks Meis3 expression in the neuroectoderm, triggering the loss of posterior neural fates. Ectopic Meis3 protein expression is sufficient to rescue this phenotype. Moreover, Wnt3a induction of the posterior nervous system requires functional Meis3 in the neural plate. Using ChIP and promoter analysis, we show that Meis3 is a direct target of Wnt/beta-catenin signaling. This suggests a new model for neural anteroposterior patterning, in which Wnt3a from the paraxial mesoderm induces posterior cell fates via direct activation of a crucial transcription factor in the overlying neural plate.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Mesodermo/embriología , Placa Neural/embriología , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo , Proteínas de Xenopus/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Inmunoprecipitación de Cromatina , Proteínas de Homeodominio/genética , Hibridación in Situ , Técnicas In Vitro , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Proteínas Wnt/genética , Proteína Wnt3 , Proteína Wnt3A , Proteínas de Xenopus/genética , Xenopus laevis , Proteínas de Pez Cebra/genética , beta Catenina/genética , beta Catenina/metabolismo
14.
RNA ; 17(5): 944-56, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21444632

RESUMEN

Post-transcriptional control of mRNA stability and translation is central to multiple developmental pathways. This control can be linked to cytoplasmic polyadenylation in certain settings. In maturing Xenopus oocytes, specific mRNAs are targeted for polyadenylation via recruitment of the Cytoplasmic Polyadenylation Element (CPE) binding protein (CPEB) to CPE(s) within the 3' UTR. Cytoplasmic polyadenylation is also critical to early embryonic events, although corresponding determinants are less defined. Here, we demonstrate that the Xenopus ortholog of the poly(rC) binding protein αCP2 can recruit cytoplasmic poly(A) polymerase activity to mRNAs in Xenopus post-fertilization embryos, and that this recruitment relies on cis sequences recognized by αCP2. We find that the hα-globin 3' UTR, a validated mammalian αCP2 target, constitutes an effective target for cytoplasmic polyadenylation in Xenopus embryos, but not during Xenopus oocyte maturation. We further demonstrate that the cytoplasmic polyadenylation activity is dependent on the action of the C-rich αCP-binding site in conjunction with the adjacent AAUAAA. Consistent with its ability to target mRNA for poly(A) addition, we find that XαCP2 associates with core components of the Xenopus cytoplasmic polyadenylation complex, including the cytoplasmic poly(A) polymerase XGLD2. Furthermore, we observe that the C-rich αCP-binding site can robustly enhance the activity of a weak canonical oocyte maturation CPE in early embryos, possibly via a direct interaction between XαCP2 and CPEB1. These studies establish XαCP2 as a novel cytoplasmic polyadenylation trans factor, indicate that C-rich sequences can function as noncanonical cytoplasmic polyadenylation elements, and expand our understanding of the complexities underlying cytoplasmic polyadenylation in specific developmental settings.


Asunto(s)
Citoplasma/metabolismo , Poli C/metabolismo , Poliadenilación , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Regiones no Traducidas 3' , Animales , Femenino , Oocitos/citología , Oocitos/metabolismo , Unión Proteica , ARN Mensajero/genética , Especificidad por Sustrato , Proteínas de Xenopus/genética , Xenopus laevis/embriología , Factores de Escisión y Poliadenilación de ARNm/genética
15.
Blood ; 118(22): 5794-8, 2011 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21881043

RESUMEN

Familial hemophagocytic lymphohistiocytosis (HLH) is a rare primary immunodeficiency disorder characterized by defects in cell-mediated cytotoxicity that results in fever, hepatosplenomegaly, and cytopenias. Familial HLH is well recognized in children but rarely diagnosed in adults. We conducted a retrospective review of genetic and immunologic test results in patients who developed HLH in adulthood. Included in our study were 1531 patients with a clinical diagnosis of HLH; 175 patients were 18 years or older. Missense and splice-site sequence variants in PRF1, MUNC13-4, and STXBP2 were found in 25 (14%) of the adult patients. The A91V-PRF1 genotype was found in 12 of these patients (48%). The preponderance of hypomorphic mutations in familial HLH-causing genes correlates with the later-onset clinical symptoms and the more indolent course in adult patients. We conclude that late-onset familial HLH occurs more commonly than was suspected previously.


Asunto(s)
Linfohistiocitosis Hemofagocítica/genética , Proteínas de la Membrana/genética , Proteínas Munc18/genética , Mutación , Proteínas Citotóxicas Formadoras de Poros/genética , Adolescente , Adulto , Edad de Inicio , Anciano , Análisis Mutacional de ADN , Femenino , Frecuencia de los Genes , Humanos , Linfohistiocitosis Hemofagocítica/epidemiología , Masculino , Proteínas de la Membrana/fisiología , Persona de Mediana Edad , Proteínas Munc18/fisiología , Mutación/fisiología , Perforina , Proteínas Citotóxicas Formadoras de Poros/fisiología , Adulto Joven
16.
Cells ; 12(18)2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37759479

RESUMEN

The Wnt signaling pathway is a highly conserved regulator of metazoan development and stem cell maintenance. Activation of Wnt signaling is an early step in diverse malignancies. Work over the past four decades has defined a "canonical" Wnt pathway that is initiated by Wnt proteins, secreted glycoproteins that bind to a surface receptor complex and activate intracellular signal transduction by inhibiting a catalytic complex composed of the classical tumor suppressor Adenomatous Polyposis Coli (APC), Axin, and Glycogen Synthase Kinase-3 (GSK-3). The best characterized effector of this complex is ß-catenin, which is stabilized by inhibition of GSK-3, allowing ß-catenin entrance to the nucleus and activation of Wnt target gene transcription, leading to multiple cancers when inappropriately activated. However, canonical Wnt signaling through the APC/Axin/GSK-3 complex impinges on other effectors, independently of ß-catenin, including the mechanistic Target of Rapamycin (mTOR), regulators of protein stability, mitotic spindle orientation, and Hippo signaling. This review focuses on these alternative effectors of the canonical Wnt pathway and how they may contribute to cancers.


Asunto(s)
Poliposis Adenomatosa del Colon , Vía de Señalización Wnt , Animales , Glucógeno Sintasa Quinasa 3 , Proteína Axina , beta Catenina
17.
J Dev Biol ; 11(3)2023 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-37489330

RESUMEN

Neural crest (NC) is a unique vertebrate cell type arising from the border of the neural plate and epidermis that gives rise to diverse tissues along the entire body axis. Roberto Mayor and colleagues have made major contributions to our understanding of NC induction, delamination, and migration. We report that a truncating mutation of the classical tumor suppressor Adenomatous Polyposis Coli (apc) disrupts craniofacial development in zebrafish larvae, with a marked reduction in the cranial neural crest (CNC) cells that contribute to mandibular and hyoid pharyngeal arches. While the mechanism is not yet clear, the altered expression of signaling molecules that guide CNC migration could underlie this phenotype. For example, apcmcr/mcr larvae express substantially higher levels of complement c3, which Mayor and colleagues showed impairs CNC cell migration when overexpressed. However, we also observe reduction in stroma-derived factor 1 (sdf1/cxcl12), which is required for CNC migration into the head. Consistent with our previous work showing that APC directly enhances the activity of glycogen synthase kinase 3 (GSK-3) and, independently, that GSK-3 phosphorylates multiple core mRNA splicing factors, we identify 340 mRNA splicing variations in apc mutant zebrafish, including a splice variant that deletes a conserved domain in semaphorin 3f (sema3f), an axonal guidance molecule and a known regulator of CNC migration. Here, we discuss potential roles for apc in CNC development in the context of some of the seminal findings of Mayor and colleagues.

18.
bioRxiv ; 2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-38077058

RESUMEN

Hematopoietic stem cell (HSC) transplantation using umbilical cord blood (UCB) is a potentially life-saving treatment for leukemia and bone marrow failure but is limited by the low number of HSCs in UCB. The loss of HSCs after ex vivo manipulation is also a major obstacle to gene editing for inherited blood disorders. HSCs require a low rate of translation to maintain their capacity for self-renewal, but hematopoietic cytokines used to expand HSCs stimulate protein synthesis and impair long-term self-renewal. We previously described cytokine-free conditions that maintain but do not expand human and mouse HSCs ex vivo. Here we performed a high throughput screen and identified translation inhibitors that allow ex vivo expansion of human HSCs while minimizing cytokine exposure. Transplantation assays show a ~5-fold expansion of long-term HSCs from UCB after one week of culture in low cytokine conditions. Single cell transcriptomic analysis demonstrates maintenance of HSCs expressing mediators of the unfolded protein stress response, further supporting the importance of regulated proteostasis in HSC maintenance and expansion. This expansion method maintains and expands human HSCs after CRISPR/Cas9 editing of the BCL11A+58 enhancer, overcoming a major obstacle to ex vivo gene correction for human hemoglobinopathies.

19.
Dev Biol ; 357(2): 478-91, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21741375

RESUMEN

Most zygotic genes remain transcriptionally silent in Drosophila, Xenopus, and zebrafish embryos through multiple mitotic divisions until the midblastula transition (MBT). Several genes have been identified in each of these organisms that are transcribed before the MBT, but whether precocious expression of specific mRNAs is important for later development has not been examined in detail. Here, we identify a class of protein coding transcripts activated before the MBT by the maternal T-box factor VegT that are components of an established transcriptional regulatory network required for mesendoderm induction in Xenopus laevis, including the Nodal related ligands xnr5, xnr6, and derrière and the transcription factors bix4, and sox17α. Accumulation of phospho-Smad2, a hallmark of active Nodal signaling, at the onset of the MBT requires preMBT transcription and activity of xnr5 and xnr6. Furthermore, preMBT activation of the Nodal pathway is essential for mesendodermal gene expression and patterning of the embryo. Finally, xnr5 and xnr6 can also activate their own expression during cleavage stages, indicating that preMBT transcription contributes to a feed-forward system that allows robust activation of Nodal signaling at the MBT.


Asunto(s)
Blástula/embriología , Blástula/metabolismo , Transcripción Genética , Xenopus laevis/embriología , Xenopus laevis/genética , Animales , Blástula/citología , Embrión no Mamífero/metabolismo , Endodermo/embriología , Retroalimentación Fisiológica , Regulación del Desarrollo de la Expresión Génica , Proteína Nodal/genética , Proteína Nodal/metabolismo , Transducción de Señal/genética , Proteínas Smad/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
20.
Bone ; 154: 116237, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34695616

RESUMEN

Mucopolysaccharidosis (MPS) I is a lysosomal storage disease characterized by deficient activity of the enzyme alpha-L-iduronidase, leading to abnormal accumulation of heparan and dermatan sulfate glycosaminoglycans in cells and tissues. Patients commonly exhibit progressive skeletal abnormalities, in part due to failures of endochondral ossification during postnatal growth. Previously, using the naturally-occurring canine model, we showed that bone and cartilage cells in MPS I exhibit elevated lysosomal storage from an early age and that animals subsequently exhibit significantly diminished vertebral trabecular bone formation. Wnts are critical regulators of endochondral ossification that depend on glycosaminoglycans for signaling. The objective of this study was to examine whether lithium, a glycogen synthase kinase-3 inhibitor and stimulator of Wnt/beta-catenin signaling, administered during postnatal growth could attenuate progression of vertebral trabecular bone disease in MPS I. MPS I dogs were treated orally with therapeutic levels of lithium carbonate from 14 days to 6 months-of-age. Untreated heterozygous and MPS I dogs served as controls. Serum was collected at 3 and 6 months for assessment of bone turnover markers. At the study end point, thoracic vertebrae were excised and assessed using microcomputed tomography and histology. Lithium-treated animals exhibited significantly improved trabecular spacing, number and connectivity density, and serum bone-specific alkaline phosphatase levels compared to untreated animals. Growth plates from lithium-treated animals exhibited increased numbers of hypertrophic chondrocytes relative to both untreated MPS I and heterozygous animals. These findings suggest that bone and cartilage cells in MPS I are still capable of responding to exogenous osteogenic signals even in the presence of significant lysosomal storage, and that targeted osteogenic therapies may represent a promising approach for attenuating bone disease progression in MPS I.


Asunto(s)
Enfermedades Óseas , Mucopolisacaridosis I , Animales , Enfermedades Óseas/terapia , Modelos Animales de Enfermedad , Perros , Humanos , Litio/uso terapéutico , Mucopolisacaridosis I/tratamiento farmacológico , Mucopolisacaridosis I/patología , Vértebras Torácicas/patología , Microtomografía por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA