Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Nat Biotechnol ; 40(10): 1500-1508, 2022 10.
Article in English | MEDLINE | ID: mdl-35654979

ABSTRACT

Therapeutics based on short interfering RNAs (siRNAs) delivered to hepatocytes have been approved, but new delivery solutions are needed to target additional organs. Here we show that conjugation of 2'-O-hexadecyl (C16) to siRNAs enables safe, potent and durable silencing in the central nervous system (CNS), eye and lung in rodents and non-human primates with broad cell type specificity. We show that intrathecally or intracerebroventricularly delivered C16-siRNAs were active across CNS regions and cell types, with sustained RNA interference (RNAi) activity for at least 3 months. Similarly, intravitreal administration to the eye or intranasal administration to the lung resulted in a potent and durable knockdown. The preclinical efficacy of an siRNA targeting the amyloid precursor protein was evaluated through intracerebroventricular dosing in a mouse model of Alzheimer's disease, resulting in amelioration of physiological and behavioral deficits. Altogether, C16 conjugation of siRNAs has the potential for safe therapeutic silencing of target genes outside the liver with infrequent dosing.


Subject(s)
Amyloid beta-Protein Precursor , RNAi Therapeutics , Animals , Mice , Primates/genetics , Primates/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use
2.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33443159

ABSTRACT

Inositol-1,4,5-triphosphate (IP3) kinase B (ITPKB) is a ubiquitously expressed lipid kinase that inactivates IP3, a secondary messenger that stimulates calcium release from the endoplasmic reticulum (ER). Genome-wide association studies have identified common variants in the ITPKB gene locus associated with reduced risk of sporadic Parkinson's disease (PD). Here, we investigate whether ITPKB activity or expression level impacts PD phenotypes in cellular and animal models. In primary neurons, knockdown or pharmacological inhibition of ITPKB increased levels of phosphorylated, insoluble α-synuclein pathology following treatment with α-synuclein preformed fibrils (PFFs). Conversely, ITPKB overexpression reduced PFF-induced α-synuclein aggregation. We also demonstrate that ITPKB inhibition or knockdown increases intracellular calcium levels in neurons, leading to an accumulation of calcium in mitochondria that increases respiration and inhibits the initiation of autophagy, suggesting that ITPKB regulates α-synuclein pathology by inhibiting ER-to-mitochondria calcium transport. Furthermore, the effects of ITPKB on mitochondrial calcium and respiration were prevented by pretreatment with pharmacological inhibitors of the mitochondrial calcium uniporter complex, which was also sufficient to reduce α-synuclein pathology in PFF-treated neurons. Taken together, these results identify ITPKB as a negative regulator of α-synuclein aggregation and highlight modulation of ER-to-mitochondria calcium flux as a therapeutic strategy for the treatment of sporadic PD.


Subject(s)
Calcium/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , alpha-Synuclein/metabolism , Animals , Autophagy/genetics , Endoplasmic Reticulum/metabolism , Genome-Wide Association Study/methods , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Neurons/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Phosphorylation/genetics , Signal Transduction/genetics , Synucleinopathies/genetics , Synucleinopathies/metabolism
3.
J Clin Invest ; 127(11): 3954-3969, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28990932

ABSTRACT

Uromodulin-associated kidney disease (UAKD) is caused by mutations in the uromodulin (UMOD) gene that result in a misfolded form of UMOD protein, which is normally secreted by nephrons. In UAKD patients, mutant UMOD is poorly secreted and accumulates in the ER of distal kidney epithelium, but its role in disease progression is largely unknown. Here, we modeled UMOD accumulation in mice by expressing the murine equivalent of the human UMOD p.Cys148Trp point mutation (UmodC147W/+ mice). Like affected humans, these UmodC147W/+ mice developed spontaneous and progressive kidney disease with organ failure over 24 weeks. Analysis of diseased kidneys and purified UMOD-producing cells revealed early activation of the PKR-like ER kinase/activating transcription factor 4 (PERK/ATF4) ER stress pathway, innate immune mediators, and increased apoptotic signaling, including caspase-3 activation. Unexpectedly, we also detected autophagy deficiency. Human cells expressing UMOD p.Cys147Trp recapitulated the findings in UmodC147W/+ mice, and autophagy activation with mTOR inhibitors stimulated the intracellular removal of aggregated mutant UMOD. Human cells producing mutant UMOD were susceptible to TNF-α- and TRAIL-mediated apoptosis due to increased expression of the ER stress mediator tribbles-3. Blocking TNF-α in vivo with the soluble recombinant fusion protein TNFR:Fc slowed disease progression in UmodC147W/+ mice by reducing active caspase-3, thereby preventing tubule cell death and loss of epithelial function. These findings reveal a targetable mechanism for disease processes involved in UAKD.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Renal Insufficiency/genetics , Uromodulin/genetics , Animals , Autophagy , Base Sequence , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Disease Progression , Female , Genetic Association Studies , Humans , Immunity, Innate , Male , Mice, Transgenic , Mutation, Missense , Point Mutation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Renal Insufficiency/immunology , Renal Insufficiency/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Uromodulin/metabolism
4.
Biomaterials ; 141: 314-329, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28711779

ABSTRACT

Loss of the microvascular (MV) network results in tissue ischemia, loss of tissue function, and is a hallmark of chronic diseases. The incorporation of a functional vascular network with that of the host remains a challenge to utilizing engineered tissues in clinically relevant therapies. We showed that vascular-bed-specific endothelial cells (ECs) exhibit differing angiogenic capacities, with kidney microvascular endothelial cells (MVECs) being the most deficient, and sought to explore the underlying mechanism. Constitutive activation of the phosphatase PTEN in kidney MVECs resulted in impaired PI3K/AKT activity in response to vascular endothelial growth factor (VEGF). Suppression of PTEN in vivo resulted in microvascular regeneration, but was insufficient to improve tissue function. Promoter analysis of the differentially regulated genes in KMVECs suggests that the transcription factor FOXO1 is highly active and RNAseq analysis revealed that hyperactive FOXO1 inhibits VEGF-Notch-dependent tip-cell formation by direct and indirect inhibition of DLL4 expression in response to VEGF. Inhibition of FOXO1 enhanced angiogenesis in human bio-engineered capillaries, and resulted in microvascular regeneration and improved function in mouse models of injury-repair.


Subject(s)
Forkhead Box Protein O1/metabolism , Kidney/blood supply , Kidney/physiopathology , Microvessels/physiopathology , Neovascularization, Physiologic , Adult , Animals , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Kidney/injuries , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Microvessels/metabolism , Microvessels/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
Development ; 144(13): 2428-2444, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28536097

ABSTRACT

The transcriptional pathways activated downstream of vascular endothelial growth factor (VEGF) signaling during angiogenesis remain incompletely characterized. By assessing the signals responsible for induction of the Notch ligand delta-like 4 (DLL4) in endothelial cells, we find that activation of the MAPK/ERK pathway mirrors the rapid and dynamic induction of DLL4 transcription and that this pathway is required for DLL4 expression. Furthermore, VEGF/ERK signaling induces phosphorylation and activation of the ETS transcription factor ERG, a prerequisite for DLL4 induction. Transcription of DLL4 coincides with dynamic ERG-dependent recruitment of the transcriptional co-activator p300. Genome-wide gene expression profiling identified a network of VEGF-responsive and ERG-dependent genes, and ERG chromatin immunoprecipitation (ChIP)-seq revealed the presence of conserved ERG-bound putative enhancer elements near these target genes. Functional experiments performed in vitro and in vivo confirm that this network of genes requires ERK, ERG and p300 activity. Finally, genome-editing and transgenic approaches demonstrate that a highly conserved ERG-bound enhancer located upstream of HLX (which encodes a transcription factor implicated in sprouting angiogenesis) is required for its VEGF-mediated induction. Collectively, these findings elucidate a novel transcriptional pathway contributing to VEGF-dependent angiogenesis.


Subject(s)
E1A-Associated p300 Protein/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Transcription, Genetic/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cattle , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation, Developmental/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Introns/genetics , MAP Kinase Signaling System/drug effects , Male , Mice , Neovascularization, Physiologic/genetics , Transcriptional Regulator ERG/metabolism , Zebrafish/embryology
6.
Blood ; 125(20): 3202-12, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25838349

ABSTRACT

The blood contains high concentrations of circulating extracellular vesicles (EVs), and their levels and contents are altered in several disease states, including cardiovascular disease. However, the function of circulating EVs, especially the microRNAs (miRNAs) that they contain, are poorly understood. We sought to determine the effect of secreted vesicles produced by quiescent endothelial cells (ECs) on monocyte inflammatory responses and to assess whether transfer of microRNAs occurs between these cells. We observed that monocytic cells cocultured (but not in contact) with ECs were refractory to inflammatory activation. Further characterization revealed that endothelium-derived EVs (EC-EVs) suppressed monocyte activation by enhancing immunomodulatory responses and diminishing proinflammatory responses. EVs isolated from mouse plasma also suppressed monocyte activation. Importantly, injection of EC-EVs in vivo repressed monocyte/macrophage activation, confirming our in vitro findings. We found that several antiinflammatory microRNAs were elevated in EC-EV-treated monocytes. In particular, miR-10a was transferred to monocytic cells from EC-EVs and could repress inflammatory signaling through the targeting of several components of the NF-κB pathway, including IRAK4. Our findings reveal that ECs secrete EVs that can modulate monocyte activation and suggest that altered EV secretion and/or microRNA content may affect vascular inflammation in the setting of cardiovascular disease.


Subject(s)
Endothelial Cells/metabolism , MicroRNAs/genetics , Monocytes/immunology , Monocytes/metabolism , Secretory Vesicles/metabolism , Cell Communication , Cell Line , Coculture Techniques , Extracellular Space , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interferon Regulatory Factors/metabolism , Lipopolysaccharides/immunology , NF-kappa B/metabolism , Signal Transduction
7.
Front Genet ; 5: 422, 2014.
Article in English | MEDLINE | ID: mdl-25540650

ABSTRACT

Cardiovascular diseases such as atherosclerosis are one of the leading causes of morbidity and mortality worldwide. The clinical manifestations of atherosclerosis, which include heart attack and stroke, occur several decades after initiation of the disease and become more severe with age. Inflammation of blood vessels plays a prominent role in atherogenesis. Activation of the endothelium by inflammatory mediators leads to the recruitment of circulating inflammatory cells, which drives atherosclerotic plaque formation and progression. Inflammatory signaling within the endothelium is driven predominantly by the pro-inflammatory transcription factor, NF-κB. Interestingly, activation of NF-κB is enhanced during the normal aging process and this may contribute to the development of cardiovascular disease. Importantly, studies utilizing mouse models of vascular inflammation and atherosclerosis are uncovering a network of noncoding RNAs, particularly microRNAs, which impinge on the NF-κB signaling pathway. Here we summarize the literature regarding the control of vascular inflammation by microRNAs, and provide insight into how these microRNA-based pathways might be harnessed for therapeutic treatment of disease. We also discuss emerging areas of endothelial cell biology, including the involvement of long noncoding RNAs and circulating microRNAs in the control of vascular inflammation.

8.
Development ; 141(23): 4610-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25359725

ABSTRACT

Maintenance of vascular integrity is required for embryogenesis and organ homeostasis. However, the gene expression programs that stabilize blood vessels are poorly understood. Here, we show that the histone methyltransferase Ezh2 maintains integrity of the developing vasculature by repressing a transcriptional program that activates expression of Mmp9. Inactivation of Ezh2 in developing mouse endothelium caused embryonic lethality with compromised vascular integrity and increased extracellular matrix degradation. Genome-wide approaches showed that Ezh2 targets Mmp9 and its activators Fosl1 and Klf5. In addition, we uncovered Creb3l1 as an Ezh2 target that directly activates Mmp9 gene expression in the endothelium. Furthermore, genetic inactivation of Mmp9 rescued vascular integrity defects in Ezh2-deficient embryos. Thus, epigenetic repression of Creb3l1, Fosl1, Klf5 and Mmp9 by Ezh2 in endothelial cells maintains the integrity of the developing vasculature, potentially linking this transcriptional network to diseases with compromised vascular integrity.


Subject(s)
Blood Vessels/embryology , Epigenetic Repression/physiology , Gene Expression Regulation, Developmental/physiology , Polycomb Repressive Complex 2/metabolism , Signal Transduction/physiology , Animals , Benzothiazoles , Blotting, Western , Chromatin Immunoprecipitation , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Primers/genetics , Diamines , Enhancer of Zeste Homolog 2 Protein , Epigenetic Repression/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , In Situ Hybridization , Kruppel-Like Transcription Factors , Luciferases , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Microscopy, Electron, Transmission , Nerve Tissue Proteins/metabolism , Organic Chemicals , Polycomb Repressive Complex 2/genetics , Proto-Oncogene Proteins c-fos/metabolism , Quinolines , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA
9.
Biomaterials ; 35(27): 7786-99, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24930852

ABSTRACT

Adhesion molecule signaling is critical to human pluripotent stem cell (hPSC) survival, self-renewal, and differentiation. Thus, hPSCs are grown as clumps of cells on feeder cell layers or poorly defined extracellular matrices such as Matrigel. We sought to define a small molecule that would initiate adhesion-based signaling to serve as a basis for a defined substrate for hPSC culture. Soluble angiopoeitin-1 (Ang-1)-derived peptide QHREDGS added to defined serum-free media increased hPSC colony cell number and size during long- and short-term culture when grown on feeder cell layers or Matrigel, i.e. on standard substrates, without affecting hPSC morphology, growth rate or the ability to differentiate into multiple lineages both in vitro and in vivo. Importantly, QHREDGS treatment decreased hPSC apoptosis during routine passaging and single-cell dissociation. Mechanistically, the interaction of QHREDGS with ß1-integrins increased expression of integrin-linked kinase (ILK), increased expression and activation of extracellular signal-regulated kinases 1/2 (ERK1/2), and decreased caspase-3/7 activity. QHREDGS immobilization to polyethylene glycol hydrogels significantly increased cell adhesion in a dose-dependent manner. We propose QHREDGS as a small molecule inhibitor of hPSC apoptosis and the basis of an affordable defined substrate for hPSC maintenance.


Subject(s)
Angiopoietin-1/pharmacology , Apoptosis/drug effects , Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/cytology , Peptides/pharmacology , Animals , Caspases/metabolism , Cell Adhesion/drug effects , Cell Count , Cell Proliferation/drug effects , Cell Size/drug effects , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Feeder Cells/cytology , Feeder Cells/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/enzymology , Integrin beta1/metabolism , Mice , Protein Serine-Threonine Kinases/metabolism , Time Factors
10.
Dev Cell ; 26(1): 45-58, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23830865

ABSTRACT

Vegf signaling specifies arterial fate during early vascular development by inducing the transcription of Delta-like 4 (Dll4), the earliest Notch ligand gene expressed in arterial precursor cells. Dll4 expression precedes that of Notch receptors in arteries, and factors that direct its arterial-specific expression are not known. To identify the transcriptional program that initiates arterial Dll4 expression, we characterized an arterial-specific and Vegf-responsive enhancer of Dll4. Our findings demonstrate that Notch signaling is not required for initiation of Dll4 expression in arteries and suggest that Notch instead functions as a maintenance factor. Importantly, we find that Vegf signaling activates MAP kinase (MAPK)-dependent E26 transformation-specific sequence (ETS) factors in the arterial endothelium to drive expression of Dll4 and Notch4. These findings identify a Vegf/MAPK-dependent transcriptional pathway that specifies arterial identity by activating Notch signaling components and illustrate how signaling cascades can modulate broadly expressed transcription factors to achieve tissue-specific transcriptional outputs.


Subject(s)
Aorta/physiology , Gene Expression Regulation, Developmental , Vascular Endothelial Growth Factor A/metabolism , Adaptor Proteins, Signal Transducing , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/metabolism , Aorta/metabolism , Binding Sites , Calcium-Binding Proteins , Endocardium/embryology , Endocardium/metabolism , Enhancer Elements, Genetic , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Organ Specificity , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor, Notch4 , Receptors, Notch/genetics , Receptors, Notch/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic , Transcriptional Regulator ERG , Vascular Endothelial Growth Factor A/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
11.
Arterioscler Thromb Vasc Biol ; 33(2): 193-200, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23325476

ABSTRACT

The regulated response of endothelial cells to signals in their environment is not only critical for the de novo formation of primordial vascular networks during early development (ie, vasculogenesis), but is also required for the subsequent growth and remodeling of new blood vessels from preexisting ones (ie, angiogenesis). Vascular endothelial growth factors (Vegfs) and their endothelial cell-specific receptors play a crucial role in nearly all aspects of blood vessel growth. How the outputs from these pathways affect and coordinate endothelial behavior is an area of intense research. Recently, numerous studies have highlighted roles for microRNAs in modulating Vegf signaling output in several different contexts. In this review, we will provide an overview of how small RNAs regulate multiple aspects of the Vegf signaling pathway. In particular, we highlight areas where identification of microRNAs and their targets has provided new insight into the role of downstream effectors in modulating Vegf output during development. As Vegf plays a broad role in multiple aspects of endothelial biology and has become a target for therapeutic manipulation of pathological blood vessel growth, microRNAs that affect Vegf signaling output will undoubtedly be major targets of clinical value.


Subject(s)
Blood Vessels/metabolism , Endothelial Cells/metabolism , MicroRNAs/metabolism , Neovascularization, Physiologic , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Blood Vessels/embryology , Gene Expression Regulation, Developmental , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Morphogenesis , Neovascularization, Physiologic/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/genetics
12.
Stem Cells Dev ; 21(15): 2838-51, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-22594450

ABSTRACT

Inducing a stable and predictable program of neural cell fate in pluripotent cells in vitro is an important goal for utilizing these cells for modeling human disease mechanisms. However, the extent to which in vitro neural specification recapitulates in vivo neural specification remains to be fully established. We previously demonstrated that in the mouse embryo, activation of fibroblast growth factor (FGF) signalling promotes definitive neural stem cell (NSC) development through the upregulation of the transcription factor Zfhx1b. Here, we asked whether Zfhx1b is similarly required during neural lineage development of embryonic stem (ES) cells. Zfhx1b gene expression is rapidly upregulated in mouse ES cells cultured in a permissive neural-inducing environment, compared to ES cells in a standard pluripotency maintenance environment, and is potentiated by FGF signalling. However, overexpression of Zfhx1b in ES cells in maintenance conditions, containing serum and leukemia inhibitory factor (LIF), is sufficient to induce Sox1 expression, a marker found in neural precursors and to promote definitive NSC colony formation. Knockdown of Zfhx1b in ES cells using siRNA did not affect the initial transition of ES cells to a neural cell fate, but did diminish the ability of these neural cells to develop further into definitive NSCs. Thus, our findings using ES cells are congruent with evidence from mouse embryos and support a model, whereby intercellular FGF signaling induces Zfhx1b, which promotes the development of definitive NSCs subsequent to an initial neural specification event that is independent of this pathway.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/physiology , Homeodomain Proteins/physiology , Neural Stem Cells/metabolism , Repressor Proteins/physiology , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Body Patterning , Cells, Cultured , Coculture Techniques , Embryonic Stem Cells/metabolism , Fibroblast Growth Factor 8/physiology , Gene Expression , Glycoproteins/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Leukemia Inhibitory Factor/physiology , Mice , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Up-Regulation , Zinc Finger E-box Binding Homeobox 2
13.
Neural Dev ; 5: 13, 2010 May 06.
Article in English | MEDLINE | ID: mdl-20459606

ABSTRACT

BACKGROUND: Mouse definitive neural stem cells (NSCs) are derived from a population of LIF-responsive primitive neural stem cells (pNSCs) within the neurectoderm, yet details on the early signaling and transcriptional mechanisms that control this lineage transition are lacking. Here we tested whether FGF and Wnt signaling pathways can regulate Zfhx1b expression to control early neural stem cell development. RESULTS: By microinjecting FGF8b into the pro-amniotic cavity ex vivo at 7.0 days post-coitum (dpc) and culturing whole embryos, we demonstrate that neurectoderm-specific gene expression (for example, Sox2, Nestin, Zfhx1b) is increased, whereas Wnt3a represses neurectoderm gene expression. To determine whether FGF signaling also mediates the lineage transition from a pNSC to a NSC, 7.0-dpc embryos were microinjected with either FGF8b or inhibitors of the FGF receptor-MAP kinase signaling pathway ex vivo, cultured as whole embryos to approximately 8.5 dpc and assayed for clonal NSC colony formation. We show that pre-activation of FGF signaling in the anterior neurectoderm causes an increase in the number of colony forming NSCs derived later from the anterior neural plate, whereas inhibition of FGF signaling significantly reduces the number of NSC colonies. Interestingly, inhibition of FGF signaling causes the persistence of LIF-responsive pNSCs within the anterior neural plate and over-expression of Zfhx1b in these cells is sufficient to rescue the transition from a LIF-responsive pNSC to an FGF-responsive NSC. CONCLUSION: Our data suggest that definitive NSC fate specification in the mouse neurectoderm is facilitated by FGF activation of Zfhx1b.


Subject(s)
Ectoderm/embryology , Fibroblast Growth Factors/genetics , Homeodomain Proteins/genetics , Nervous System/embryology , Neurogenesis/genetics , Repressor Proteins/genetics , Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/drug effects , Cell Lineage/genetics , Cell Proliferation/drug effects , Cells, Cultured , Colony-Forming Units Assay , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/pharmacology , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Neural Tube/embryology , Neurogenesis/drug effects , Neurons/cytology , Neurons/metabolism , Receptors, Fibroblast Growth Factor/agonists , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Stem Cells/cytology , Stem Cells/drug effects , Wnt Proteins/genetics , Wnt Proteins/pharmacology , Wnt3 Protein , Wnt3A Protein , Zinc Finger E-box Binding Homeobox 2
SELECTION OF CITATIONS
SEARCH DETAIL