Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Cereb Cortex ; 30(12): 6444-6457, 2020 11 03.
Article in English | MEDLINE | ID: mdl-32780108

ABSTRACT

The development of the cerebral cortex is directed by a series of methodically precise events, including progenitor cell proliferation, neural differentiation, and cell positioning. Over the past decade, many studies have demonstrated the critical contributions of Notch signaling in neurogenesis, including that in the developing telencephalon. However, in vivo evidence for the role of Notch signaling in cortical development still remains limited partly due to the redundant functions of four mammalian Notch paralogues and embryonic lethality of the knockout mice. Here, we utilized the conditional deletion and in vivo gene manipulation of Rbpj, a transcription factor that mediates signaling by all four Notch receptors, to overcome these challenges and examined the specific roles of Rbpj in cortical development. We report severe structural abnormalities in the embryonic and postnatal cerebral cortex in Rbpj conditional knockout mice, which provide strong in vivo corroboration of previously reported functions of Notch signaling in neural development. Our results also provide evidence for a novel dual role of Rbpj in cell type-specific regulation of two key developmental events in the cerebral cortex: the maintenance of the undifferentiated state of neural progenitor cells, and the radial and tangential allocation of neurons, possibly through stage-dependent differential regulation of Ngn1.


Subject(s)
Cell Movement , Cerebral Cortex/growth & development , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Neural Stem Cells/physiology , Neurons/physiology , Animals , Cell Differentiation , Cerebral Cortex/cytology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/cytology
2.
Cell Mol Life Sci ; 76(5): 921-940, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30470852

ABSTRACT

The transcription factor Ptf1a is a crucial helix-loop-helix (bHLH) protein selectively expressed in the pancreas, retina, spinal cord, brain, and enteric nervous system. Ptf1a is preferably assembled into a transcription trimeric complex PTF1 with an E protein and Rbpj (or Rbpjl). In pancreatic development, Ptf1a is indispensable in controlling the expansion of multipotent progenitor cells as well as the specification and maintenance of the acinar cells. In neural tissues, Ptf1a is transiently expressed in the post-mitotic cells and specifies the inhibitory neuronal cell fates, mostly mediated by downstream genes such as Tfap2a/b and Prdm13. Mutations in the coding and non-coding regulatory sequences resulting in Ptf1a gain- or loss-of-function are associated with genetic diseases such as pancreatic and cerebellar agenesis in the rodent and human. Surprisingly, Ptf1a alone is sufficient to reprogram mouse or human fibroblasts into tripotential neural stem cells. Its pleiotropic functions in many biological processes remain to be deciphered in the future.


Subject(s)
Cellular Reprogramming , Transcription Factors/physiology , Animals , Brain/embryology , Cell Transdifferentiation , Enteric Nervous System/embryology , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Pancreas/embryology , Pancreas/physiology , Retina/embryology , Spinal Cord/embryology , Transcription Factors/chemistry , Transcription Factors/genetics
3.
J Neurosci ; 38(21): 5022-5037, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29739871

ABSTRACT

The adult olfactory epithelium (OE) has the remarkable capacity to regenerate fully both neurosensory and non-neuronal cell types after severe epithelial injury. Lifelong persistence of two stem cell populations supports OE regeneration when damaged: the horizontal basal cells (HBCs), dormant and held in reserve; and globose basal cells, a heterogeneous population most of which are actively dividing. Both populations regenerate all cell types of the OE after injury, but the mechanisms underlying neuronal versus non-neuronal lineage commitment after recruitment of the stem cell pools remains unknown. We used both retroviral transduction and mouse lines that permit conditional cell-specific genetic manipulation as well as the tracing of progeny to study the role of canonical Notch signaling in the determination of neuronal versus non-neuronal lineages in the regenerating adult OE. Excision of either Notch1 or Notch2 genes alone in HBCs did not alter progenitor fate during recovery from epithelial injury, whereas conditional knock-out of both Notch1 and Notch2 together, retroviral transduction of progenitors with a dominant-negative form of MAML (mastermind-like), or excision of the downstream cofactor RBPJ caused progeny to adopt a neuronal fate exclusively. Conversely, we show that overexpressing the Notch1-intracellular domain (N1ICD) either genetically or by transduction blocks neuronal differentiation completely. However, N1ICD overexpression requires both alleles of the canonical cofactor RBPJ to specify downstream lineage. Together, our results suggest that canonical RBPJ-dependent Notch signaling through redundant Notch1 and Notch2 receptors is both necessary and sufficient for determining neuronal versus non-neuronal differentiation in the regenerating adult OE.SIGNIFICANCE STATEMENT Despite the substantial reconstitution of the olfactory epithelium and its population of sensory neurons after injury, disruption and exhaustion of neurogenesis is a consequence of aging and a cause of olfactory dysfunction. Understanding the mechanisms underlying the generation of replacement neurons and non-neuronal cells is critical to any therapeutic strategy aimed at rebuilding a functional neuroepithelium. The results shown here demonstrate that canonical Notch signaling determines the balance between neurons and non-neuronal cells during restoration of the epithelium after injury. Moreover, the complexities of the multiple Notch pathways impinging on that decision are dissected in detail. Finally, RBPJ, the canonical Notch transcriptional cofactor, exhibits a heretofore unreported haploinsufficiency in setting the balance among the regenerating populations.


Subject(s)
Neural Stem Cells/physiology , Olfactory Mucosa/physiology , Receptors, Notch/physiology , Signal Transduction/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis/genetics , Neurogenesis/physiology , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Olfactory Mucosa/cytology , Rats , Rats, Sprague-Dawley , Receptor, Notch1/genetics , Receptor, Notch1/physiology , Receptor, Notch2/genetics , Receptor, Notch2/physiology , Transcription Factors/genetics , Transcription Factors/physiology
4.
Genesis ; 56(6-7): e23215, 2018 06.
Article in English | MEDLINE | ID: mdl-30134068

ABSTRACT

The neural crest-derived ensheathing glial cells of the olfactory nerve (OECs) are unique in spanning both the peripheral and central nervous systems: they ensheathe bundles of axons projecting from olfactory receptor neurons in the nasal epithelium to their targets in the olfactory bulb. OECs are clinically relevant as a promising autologous cell transplantation therapy for promoting central nervous system repair. They are also important for fertility, being required for the migration of embryonic gonadotropin-releasing hormone (GnRH) neurons from the olfactory placode along terminal nerve axons to the medial forebrain, which they enter caudal to the olfactory bulbs. Like Schwann cell precursors, OEC precursors associated with the developing olfactory nerve express the glial marker myelin protein zero and the key peripheral glial transcription factor Sox10. The transition from Schwann cell precursors to immature Schwann cells is accelerated by canonical Notch signaling via the Rbpj transcription factor. Here, we aimed to test the role of Notch/Rbpj signaling in developing OECs by blocking the pathway in both chicken and mouse. Our results suggest that Notch/Rbpj signaling prevents the cranial neural crest cells that colonize the olfactory nerve from differentiating as neurons, and at later stages contributes to the guidance of GnRH neurons.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Neural Crest/metabolism , Receptors, Notch/physiology , Animals , Cell Differentiation/physiology , Cell Movement/physiology , Chick Embryo , Gonadotropin-Releasing Hormone , Mice , Neural Crest/embryology , Neurogenesis/physiology , Neuroglia/physiology , Neurons/metabolism , Olfactory Bulb/physiology , Signal Transduction/physiology
5.
Hepatology ; 64(4): 1302-1316, 2016 10.
Article in English | MEDLINE | ID: mdl-27362333

ABSTRACT

UNLABELLED: Liver vasculature is crucial for adequate hepatic functions. Global deletion of Notch signaling in mice results in liver vascular pathologies. However, whether Notch in endothelium is essential for hepatic vascular structure and function remains unknown. To uncover the function of endothelial Notch in the liver, we deleted Rbpj, a transcription factor mediating all canonical Notch signaling, or Notch1 from the endothelium of postnatal mice. We investigated the hepatic vascular defects in these mutants. The liver was severely affected within 2 weeks of endothelial deletion of Rbpj from birth. Two-week old mutant mice had enlarged vessels on the liver surface, abnormal vascular architecture, and dilated sinusoids. Vascular casting and fluorosphere passage experiments indicated the presence of porto-systemic shunts. These mutant mice presented with severely necrotic liver parenchyma and significantly larger hypoxic areas, likely resulting from vascular shunts. We also found elevated levels of VEGF receptor 3 together with reduced levels of ephrin-B2, suggesting a possible contribution of these factors to the generation of hepatic vascular abnormalities. Deletion of Rbpj from the adult endothelium also led to dilated sinusoids, vascular shunts, and necrosis, albeit milder than that observed in mice with deletion from birth. Similar to deletion of Rbpj, loss of endothelial Notch1 from birth led to similar hepatic vascular malformations within 2 weeks. CONCLUSIONS: Endothelial Notch signaling is essential for the development and maintenance of proper hepatic vascular architecture and function. These findings may elucidate the molecular pathogenesis of hepatic vascular malformation and the safety of therapeutics inhibiting Notch. (Hepatology 2016;64:1302-1316).


Subject(s)
Liver/blood supply , Receptor, Notch1/physiology , Vascular Malformations/etiology , Animals , Endothelium, Vascular , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Mice , Signal Transduction
6.
Prostate ; 76(2): 140-50, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26437743

ABSTRACT

BACKGROUND: Aberrant signaling pathways leads to cancer initiation and progression. Both Notch and PI3K/AKT signaling pathways are believed to be involved in prostate cancer. How the interaction between the two pathways contributes to prostate cancer progression to androgen independence is still elusive. METHODS: Prostate cancer cells were grown in RPMI 1,640 supplemented with 10% heat-inactivated fetal bovine serum (FBS) or 10% charcoal-stripped heat-inactivated fetal bovine serum (FCS), 1% penicillin-streptomycin in 75 cm2 polystyrene flasks, and maintained at 37 °C in a humidified atmosphere with 5% CO2 . Cell proliferation, invasion were performed with cell counting, matrigel assay in vitro. Dual luciferase assays were performed using reporter plasmids with ARE (Androgen Response Element, ARE). RNA interference was applied to gene silence. Tumorigenicity of cancer cells was evaluated by mouse xenograft in vivo. RESULTS: A subpopulation of casodex resistant prostate cancer cells were identified with an overexpressed androgen receptor (AR) and aggressive phenotypes, characterized with high proliferation, invasion in vitro and enhanced tumorigenesis in vivo. Gene profiling for androgen-dependent LNCaP and androgen-independent LNCaP-CR revealed that both CSL and AKT gave the similar expressional pattern upon casodex treatment. Immunoblot demonstrated that CSL and AKT were dramatically suppressed in androgen dependent LNCaP cells, but slightly inhibited in LNCaP-CR cells as well as other androgen independent prostate cancer cells. Further studies indicated that CSL regulates AKT, and subsequently AR in prostate cancer cells. AKT mediates casodex resistance and androgen independence through regulation of cyclin D1. CONCLUSION: CSL-AKT-AR axis might play an important role in prostate cancer progression. Targeting CSL depleted the casodex resistant population through inhibition of the AKT, suggesting a more effective therapeutic strategy for abrogating casodex resistance in advanced prostate cancer.


Subject(s)
Disease Progression , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/physiology , Receptors, Androgen/physiology , Androgen Antagonists/pharmacology , Anilides/pharmacology , Animals , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, Nude , Nitriles/pharmacology , Tosyl Compounds/pharmacology
7.
J Immunol ; 191(4): 1916-26, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23833236

ABSTRACT

Although accumulation of dendritic cell (DC) precursors occurs in bone marrow, the terminal differentiation of these cells takes place outside bone marrow. The signaling, regulating this process, remains poorly understood. We demonstrated that this process could be differentially regulated by Notch ligands: Jagged-1 (Jag1) and Delta-like ligand 1 (Dll1). In contrast to Dll1, Jag1, in vitro and during induced myelopoiesis in vivo, prevented DC differentiation by promoting the accumulation of their precursors. Although both ligands activated Notch in hematopoietic progenitor cells, they had an opposite effect on Wnt signaling. Dll1 activated Wnt pathways, whereas Jag1 inhibited it via downregulation of the expression of the Wnt receptors Frizzled (Fzd). Jag1 suppressed fzd expression by retaining histone deacetylase 1 in the complex with the transcription factor CSL/CBF-1 on the fzd promoter. Our results suggest that DC differentiation, during induced myelopoiesis, can be regulated by the nature of the Notch ligand expressed on adjacent stroma cells.


Subject(s)
Dendritic Cells/cytology , Myelopoiesis/physiology , Wnt Signaling Pathway , Adoptive Transfer , Animals , Animals, Congenic , Bone Marrow Cells , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Dendritic Cells/classification , Down-Regulation , Female , Frizzled Receptors/genetics , Frizzled Receptors/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Histone Deacetylase 1/physiology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/physiology , Jagged-1 Protein , Ligands , Lymph Nodes/cytology , Membrane Proteins/deficiency , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes , Myelopoiesis/drug effects , Poly I-C/pharmacology , RNA Interference , Radiation Chimera , Receptor, Notch1/physiology , Recombinant Fusion Proteins/metabolism , Serrate-Jagged Proteins , Signal Transduction/drug effects , Spleen/cytology , Stromal Cells/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/deficiency , beta Catenin/physiology
8.
J Am Soc Nephrol ; 25(4): 773-83, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24480830

ABSTRACT

Neointima formation causes the failure of 60% of arteriovenous fistulas (AVFs) within 2 years. Neointima-forming mechanisms are controversial but possibly linked to excess proinflammatory responses and dysregulated Notch signaling. To identify how AVFs fail, we anastomosed the carotid artery to the internal jugular vein in normal and uremic mice and compared these findings with those in failed AVFs from patients with ESRD. Endothelial cells (ECs) of AVFs in uremic mice or patients expressed mesenchymal markers (FSP-1 and/or α-SMA) and exhibited increased expression and nuclear localization of Notch intracellular domain compared with ECs of AVFs in pair-fed control mice. Furthermore, expression of VE-Cadherin decreased, whereas expression of Notch1 and -4, Notch ligands, the downstream transcription factor of Notch, RBP-Jκ, and Notch target genes increased in ECs of AVFs in uremic mice. In cultured ECs, ectopic expression of Notch ligand or treatment with TGF-ß1 triggered the expression of mesenchymal markers and induced endothelial cell barrier dysfunction, both of which were blocked by Notch inhibition or RBP-Jκ knockout. Furthermore, Notch-induced defects in barrier function, invasion of inflammatory cells, and neointima formation were suppressed in mice with heterozygous knockdown of endothelial-specific RBP-Jκ. These results suggest that increased TGF-ß1, a complication of uremia, activates Notch in endothelial cells of AVFs, leading to accelerated neointima formation and AVF failure. Suppression of Notch activation could be a strategy for improving AFV function in uremia.


Subject(s)
Arteriovenous Shunt, Surgical/adverse effects , Endothelial Cells/physiology , Receptors, Notch/physiology , Renal Insufficiency, Chronic/physiopathology , Actins/analysis , Aged , Animals , Calcium-Binding Proteins/analysis , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Male , Mice , Middle Aged , Neointima , Renal Insufficiency, Chronic/pathology , S100 Calcium-Binding Protein A4 , Signal Transduction , Transforming Growth Factor beta1/physiology
9.
Dev Biol ; 380(2): 222-32, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23707899

ABSTRACT

The node triggers formation of the left-right axis in mouse embryos by establishing local asymmetry of Nodal and Cerl2 expression. We found that Wnt3 is expressed in perinodal crown cells preferentially on the left side. The enhancer responsible for Wnt3 expression was identified and found to be regulated by Foxa2 and Rbpj under the control of Notch signaling. Rbpj binding sites suppress enhancer activity in pit cells of the node, thereby ensuring crown cell-specific expression. In addition, we found that the expression of Gdf1 and Cerl2 is also regulated by Notch signaling, suggesting that such signaling may induce the expression of genes related to left-right asymmetry as a set. Furthermore, Cerl2 expression became symmetric in response to inhibition of Wnt-ß-catenin signaling. Our results suggest that Wnt signaling regulates the asymmetry of Cerl2 expression, which likely generates a left-right difference in Nodal activity at the node for further amplification in lateral plate mesoderm.


Subject(s)
Body Patterning , Wnt Signaling Pathway/physiology , Animals , Female , Hepatocyte Nuclear Factor 3-beta/physiology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Intercellular Signaling Peptides and Proteins/physiology , Mice , Mice, Inbred ICR , Wnt3 Protein/physiology
10.
Arthritis Rheum ; 65(10): 2623-33, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23839930

ABSTRACT

OBJECTIVE: Osteoarthritis (OA) is a degenerative disease resulting in severe joint cartilage destruction and disability. While the mechanisms underlying the development and progression of OA are poorly understood, gene mutations have been identified within cartilage-related signaling molecules, implicating impaired cell signaling in OA and joint disease. The Notch pathway has recently been identified as a crucial regulator of growth plate cartilage development, and components are expressed in joint tissue. This study was undertaken to investigate a novel role for Notch signaling in joint cartilage development, maintenance, and the pathogenesis of joint disease in a mouse model. METHODS: We performed the first mouse gene study in which the core Notch signaling component, RBP-Jκ, was tissue specifically deleted within joints. The Prx1Cre transgene removed Rbpjk loxP-flanked alleles in mesenchymal joint precursor cells, while the Col2Cre(ERT2) transgene specifically deleted Rbpjk in postnatal chondrocytes. Murine articular chondrocyte cultures were also used to examine Notch regulation of gene expression. RESULTS: Loss of Notch signaling in mesenchymal joint precursor cells did not affect embryonic joint development in mice, but rather, resulted in an early, progressive OA-like pathology. Additionally, partial loss of Notch signaling in murine postnatal cartilage resulted in progressive joint cartilage degeneration and an age-related OA-like pathology. Inhibition of Notch signaling altered the expression of the extracellular matrix (ECM)-related factors type II collagen (COL2A1), proteoglycan 4, COL10A1, matrix metalloproteinase 13, and ADAMTS. CONCLUSION: Our findings indicate that the RBP-Jκ-dependent Notch pathway is a novel pathway involved in joint maintenance and articular cartilage homeostasis, a critical regulator of articular cartilage ECM-related molecules, and a potentially important therapeutic target for OA-like joint disease.


Subject(s)
Cartilage, Articular/physiology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Joints/physiology , Receptors, Notch/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/physiology , Collagen Type I, alpha 1 Chain , Collagen Type II/genetics , Collagen Type II/physiology , Homeostasis/physiology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, Inbred Strains , Mice, Transgenic , Models, Animal , Osteoarthritis/physiopathology
11.
J Exp Med ; 204(7): 1653-64, 2007 Jul 09.
Article in English | MEDLINE | ID: mdl-17591855

ABSTRACT

Signaling through Notch receptors and their transcriptional effector RBP-J is essential for lymphocyte development and function, whereas its role in other immune cell types is unclear. We tested the function of the canonical Notch-RBP-J pathway in dendritic cell (DC) development and maintenance in vivo. Genetic inactivation of RBP-J in the bone marrow did not preclude DC lineage commitment but caused the reduction of splenic DC fraction. The inactivation of RBP-J in DCs using a novel DC-specific deleter strain caused selective loss of the splenic CD8(-) DC subset and reduced the frequency of cytokine-secreting CD8(-) DCs after challenge with Toll-like receptor ligands. In contrast, other splenic DC subsets and DCs in the lymph nodes and tissues were unaffected. The RBP-J-deficient splenic CD8(-) DCs were depleted at the postprogenitor stage, exhibited increased apoptosis, and lost the expression of the Notch target gene Deltex1. In the spleen, CD8(-) DCs were found adjacent to cells expressing the Notch ligand Delta-like 1 in the marginal zone (MZ). Thus, canonical Notch-RBP-J signaling controls the maintenance of CD8(-) DCs in the splenic MZ, revealing an unexpected role of the Notch pathway in the innate immune system.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Spleen/immunology , Animals , CD11c Antigen/genetics , Dendritic Cells/drug effects , Flow Cytometry , Homeostasis , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Integrases/genetics , Lipopolysaccharides/pharmacology , Mice , Mice, Transgenic , Polymerase Chain Reaction , Receptors, Notch/physiology , Signal Transduction , Toll-Like Receptors/physiology
12.
J Hepatol ; 59(1): 124-30, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23500150

ABSTRACT

BACKGROUND & AIMS: Repair from biliary damages requires the biliary specification of hepatic progenitor cells and the remodeling of ductular reactive structures into branching biliary tubules. We hypothesized that the morphogenetic role of Notch signaling is maintained during the repair process and have addressed this hypothesis using pharmacologic and genetic models of defective Notch signaling. METHODS: Treatment with DDC (3,5-diethoxycarbonyl-1,4-dihydrocollidine) or ANIT (alpha-naphthyl-isothiocyanate) was used to induce biliary damage in wild type mice and in mice with a liver specific defect in the Notch-2 receptor (Notch-2-cKO) or in RPB-Jk. Hepatic progenitor cells, ductular reaction, and mature ductules were quantified using K19 and SOX-9. RESULTS: In DDC treated wild type mice, pharmacologic Notch inhibition with dibenzazepine decreased the number of both ductular reaction and hepatic progenitor cells. Notch-2-cKO mice treated with DDC or ANIT accumulated hepatic progenitor cells that failed to progress into mature ducts. In RBP-Jk-cKO mice, mature ducts and hepatic progenitor cells were both significantly reduced with respect to similarly treated wild type mice. The mouse progenitor cell line BMOL cultured on matrigel, formed a tubular network allowing the study of tubule formation in vitro; γ-secretase inhibitor treatment and siRNAs silencing of Notch-1, Notch-2 or Jagged-1 significantly reduced both the length and number of tubular branches. CONCLUSIONS: These data demonstrate that Notch signaling plays an essential role in biliary repair. Lack of Notch-2 prevents biliary tubule formation, both in vivo and in vitro. Lack of RBP-Jk inhibits the generation of biliary-committed precursors and tubule formation.


Subject(s)
Bile Ducts, Intrahepatic/injuries , Bile Ducts, Intrahepatic/physiopathology , Receptor, Notch2/physiology , 1-Naphthylisothiocyanate/toxicity , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Bile Ducts, Intrahepatic/pathology , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/physiology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Jagged-1 Protein , Liver Regeneration/drug effects , Liver Regeneration/physiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis/drug effects , Morphogenesis/physiology , Pyridines/toxicity , RNA, Small Interfering/genetics , Receptor, Notch2/deficiency , Receptor, Notch2/genetics , Serrate-Jagged Proteins , Signal Transduction/drug effects , Stem Cells/drug effects , Stem Cells/pathology , Stem Cells/physiology
13.
Circ Res ; 108(7): 824-36, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21311046

ABSTRACT

RATIONALE: The proepicardium is a transient structure comprising epicardial progenitor cells located at the posterior limit of the embryonic cardiac inflow. A network of signals regulates proepicardial cell fate and defines myocardial and nonmyocardial domains at the venous pole of the heart. During cardiac development, epicardial-derived cells also contribute to coronary vessel morphogenesis. OBJECTIVE: To study Notch function during proepicardium development and coronary vessel formation in the mouse. METHODS AND RESULTS: Using in situ hybridization, RT-PCR, and immunohistochemistry, we find that Notch pathway elements are differentially activated throughout the proepicardial-epicardial-coronary transition. Analysis of RBPJk-targeted embryos indicates that Notch ablation causes ectopic procardiogenic signaling in the proepicardium that in turn promotes myocardial differentiation in adjacent mesodermal progenitors, resulting in a premature muscularization of the sinus venosus horns. Epicardium-specific Notch1 ablation using a Wt1-Cre driver line disrupts coronary artery differentiation, reduces myocardium wall thickness and myocyte proliferation, and reduces Raldh2 expression. Ectopic Notch1 activation disrupts epicardium development and causes thinning of ventricular walls. CONCLUSIONS: Epicardial Notch modulates cell differentiation in the proepicardium and adjacent pericardial mesoderm. Notch1 is later required for arterial endothelium commitment and differentiation and for vessel wall maturation during coronary vessel development and myocardium growth.


Subject(s)
Blood Circulation/physiology , Coronary Vessels/embryology , Morphogenesis/physiology , Pericardium/embryology , Receptors, Notch/physiology , Signal Transduction/physiology , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/physiology , Animals , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/physiology , Cell Differentiation/physiology , Cell Proliferation , Coronary Vessels/cytology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Mice , Mice, Inbred Strains , Mice, Transgenic , Models, Animal , Mutation , Pericardium/cytology , Receptor, Notch1/genetics , Receptor, Notch1/physiology , Receptors, Notch/genetics
14.
Circ Res ; 108(7): 813-23, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21252157

ABSTRACT

RATIONALE: The embryonic epicardium plays a crucial role in the formation of the coronary vasculature and in myocardial development, yet the exact contribution of epicardium-derived cells (EPDCs) to the vascular and connective tissue of the heart, and the factors that regulate epicardial differentiation, are insufficiently understood. OBJECTIVE: To define the role of Notch signaling in murine epicardial development. METHODS AND RESULTS: Using in situ hybridization and RT-PCR analyses, we detected expression of a number of Notch receptor and ligand genes in early epicardial development, as well as during formation of coronary arteries. Mice with epicardial deletion of Rbpj, the unique intracellular mediator of Notch signaling, survived to adulthood and exhibited enlarged coronary venous and arterial beds. Using a Tbx18-based genetic lineage tracing system, we show that EPDCs give rise to fibroblasts and coronary smooth muscle cells (SMCs) but not to endothelial cells in the wild type, whereas in Rbpj-deficient embryos EPDCs form and surround the developing arteries but fail to differentiate into SMCs. Conditional activation of Notch signaling results in premature SMC differentiation of epicardial cells and prevents coronary angiogenesis. We further show that Notch signaling regulates, and cooperates with transforming growth factor ß signaling in SM differentiation of EPDCs. CONCLUSIONS: Notch signaling is a crucial regulator of SM differentiation of EPDCs, and thus, of formation of a functional coronary system.


Subject(s)
Cell Differentiation/physiology , Myocytes, Smooth Muscle/cytology , Pericardium/cytology , Receptors, Notch/physiology , Signal Transduction/physiology , Animals , Atherosclerosis/physiopathology , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/physiopathology , Female , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Male , Mice , Mice, Knockout , Models, Animal , Pericardium/embryology , Pericardium/physiology , Receptor, Platelet-Derived Growth Factor beta/physiology , Receptors, Notch/genetics , Transforming Growth Factor beta/physiology
15.
J Neurosci ; 31(22): 8046-58, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21632926

ABSTRACT

The mammalian organ of Corti consists of a highly organized array of hair cells and supporting cells that originate from a common population of prosensory progenitors. Proper differentiation of this complex cellular mosaic requires lateral inhibition mediated by Notch signaling. Several studies have implicated Notch signaling in the earlier induction of the prosensory domain that lies along the length of the cochlear duct, and which forms before the onset of hair cell and supporting cell differentiation. To investigate the role of Notch signaling in prosensory domain formation, we conditionally inactivated the transcriptional mediator of canonical Notch signaling, RBPjκ, throughout the inner ear. Although RBPjκ mutants have severe vestibular defects and a shortened cochlear duct, markers of the prosensory domain appear at the normal time and location in the cochlea of RBPjκ mutants. Despite the lack of RBPjκ, hair cell and supporting cell markers also appear at appropriate times in the cochlea, suggesting that RBPjκ is dispensable for differentiation of the cochlear sensory epithelium. However, we also observed that differentiating hair cells and supporting cells rapidly die in RBPjκ mutants, suggesting a requirement of RBPjκ for cell survival in this tissue. Finally, in contrast to the chick basilar papilla, ectopic activation of Notch signaling did not induce ectopic sensory patches in nonsensory regions of the cochlea. Our results indicate that canonical Notch signaling is not necessary for prosensory specification in the mouse cochlea, suggesting that other signaling pathways may specify this highly derived sensory organ.


Subject(s)
Cochlea/growth & development , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Neurogenesis , Receptors, Notch/metabolism , Signal Transduction/genetics , Animals , Cells, Cultured , Cochlea/anatomy & histology , Cochlea/metabolism , Ear, Inner/anatomy & histology , Ear, Inner/growth & development , Ear, Inner/metabolism , Gene Expression Regulation, Developmental/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, Mutant Strains , Neurogenesis/genetics , Organ of Corti/growth & development , Organ of Corti/metabolism
16.
Dev Biol ; 360(1): 123-31, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21963536

ABSTRACT

The Notch signaling pathway regulates metazoan development, in part, by directly controlling the transcription of target genes. For a given cellular context, however, only subsets of the known target genes are transcribed when the pathway is activated. Thus, there are context-dependent mechanisms that selectively maintain repression of target gene transcription when the Notch pathway is activated. This review focuses on molecular mechanisms that have been recently reported to mediate selective repression of Notch pathway target gene transcription. These mechanisms are essential for generating the complex spatial and temporal expression patterns of Notch target genes during development.


Subject(s)
Receptors, Notch/genetics , Receptors, Notch/physiology , Animals , Binding Sites , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Models, Biological , Proteomics , Repressor Proteins/genetics , Repressor Proteins/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Transcription, Genetic
17.
Dev Biol ; 356(2): 337-49, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21624358

ABSTRACT

We have shown previously that during branching morphogenesis of the mouse prostate gland, Bone morphogenetic protein 7 functions to restrict Notch1-positive progenitor cells to the tips of the prostate buds. Here, we employed prostate-specific murine bi-genic systems to investigate the effects of gain and loss of Notch function during prostate development. We show that Nkx3.1(Cre) and Probasin(Cre) alleles drive expression of Cre recombinase to the prostate epithelium and periepithelial stroma. We investigated the effects of gain of Notch function using the Rosa(NI1C) conditional allele, which carries a constitutively active intracellular domain of Notch1 receptor. We carried out the analysis of loss of Notch function in Nkx3.1(Cre/+);RBP-J(flox/flox) prostates, where RBP-J is a ubiquitous transcriptional mediator of Notch signaling. We found that gain of Notch function resulted in inhibition of the tumor suppressor PTEN, and increase in cell proliferation and progenitor cells in the basal epithelium and smooth muscle compartments. In turn, loss of Notch/RBP-J function resulted in decreased cell proliferation and loss of epithelial and smooth muscle progenitors. Gain of Notch function resulted in an early onset of benign prostate hyperplasia by three months of age. Loss of Notch function also resulted in abnormal differentiation of the prostate epithelium and stroma. In particular, loss of Notch signaling and increase in PTEN promoted a switch from myoblast to fibroblast lineage, and a loss of smooth muscle. In summary, we show that Notch signaling is necessary for terminal differentiation of the prostate epithelium and smooth muscle, and that during normal prostate development Notch/PTEN pathway functions to maintain patterned progenitors in the epithelial and smooth muscle compartments. In addition, we found that both positive and negative modulation of Notch signaling results in abnormal organization of the prostate tissue, and can contribute to prostate disease in the adult organ.


Subject(s)
Muscle, Smooth/embryology , Prostate/embryology , Prostate/metabolism , Signal Transduction , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Epithelium/embryology , Epithelium/metabolism , Female , Homeodomain Proteins/physiology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Male , Mice , PTEN Phosphohydrolase/physiology , Phosphorylation , Prostate/cytology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Notch/physiology , Transcription Factors/physiology
18.
Dev Biol ; 353(2): 367-79, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21420948

ABSTRACT

The vertebrate inner ear contains multiple sensory patches comprised of hair cells and supporting cells. During development these sensory patches arise from prosensory cells that are specified and maintained through the expression of specific molecular factors. Disruption of Jagged1-mediated notch signaling causes a loss of some sensory patches and disruptions in others, indicating a role in some aspect of prosensory development. However, the presence of some sensory patches suggests that some level of notch activity persists in the absence of Jagged1. Therefore, the transcription factor Rbpj, which is required for nearly all notch function, was deleted in the developing otocyst. Results indicate a nearly complete absence of all prosensory patches in the inner ear with remaining hair cells located predominantly in the extreme apex of the cochlea. However, early markers of prosensory cells are still present in Rbpj-mutants, suggesting that maintenance, rather than induction, of prosensory development is dependent on notch signaling. Moreover, analysis of developing cochleae in Rbpj-mutants indicates changes in the spatiotemporal patterns of expression for p27(kip1), Atoh1 and hair cell differentiation markers implicating notch signaling in the regulation of the timing of cellular differentiation and/or in the maintenance of a stem/progenitor cell stage. Finally, the absence of Rbpj caused increased cell death in the cochlea beginning at E12. These results suggest important roles for Rbpj and notch signaling in multiple aspects of inner ear development including prosensory cell maturation, cellular differentiation and survival.


Subject(s)
Ear, Inner/embryology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Count , Cell Death , Cell Differentiation , Cochlea/cytology , Cochlea/embryology , Cochlea/metabolism , Cyclin-Dependent Kinase Inhibitor p27/deficiency , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Ear, Inner/abnormalities , Ear, Inner/cytology , Ear, Inner/metabolism , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Mutation , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Notch/metabolism , Signal Transduction
19.
J Biol Chem ; 286(17): 14892-902, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21372128

ABSTRACT

The Notch pathway is a conserved cell-to-cell signaling mechanism that mediates cell fate decisions in metazoans. Canonical signaling results in changes in gene expression, which is regulated by the nuclear effector of the pathway CSL (CBF1/RBP-J, Su(H), Lag-1). CSL is a DNA binding protein that functions as either a repressor or an activator of transcription, depending upon whether it is complexed by transcriptional corepressor or coactivator proteins, respectively. In stark contrast to CSL-coactivator complexes, e.g. the transcriptionally active CSL-Notch-Mastermind ternary complex, the structure and function of CSL-corepressor complexes are poorly understood. The corepressor MINT (Msx2-interacting nuclear target protein) has been shown in vivo to antagonize Notch signaling and shown in vitro to biochemically interact with CSL; however, the molecular details of this interaction are only partially defined. Here, we provide a quantitative thermodynamic binding analysis of CSL-MINT complexes. Using isothermal titration calorimetry, we demonstrate that MINT forms a high affinity complex with CSL, and we also delineate the domains of MINT and CSL that are necessary and sufficient for complex formation. Moreover, we show in cultured cells that this region of MINT can inhibit Notch signaling in transcriptional reporter assays. Taken together, our results provide functional insights into how CSL is converted from a repressor to an activator of transcription.


Subject(s)
Gene Expression Regulation , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Nuclear Proteins/metabolism , Receptors, Notch/genetics , Thermodynamics , Transcription, Genetic , Animals , Cells, Cultured , DNA-Binding Proteins , Fibroblasts/chemistry , Fibroblasts/cytology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/physiology , Protein Binding , RNA-Binding Proteins , Receptors, Notch/antagonists & inhibitors , Repressor Proteins , Trans-Activators
20.
Blood ; 116(25): 5443-54, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-20829372

ABSTRACT

The Notch pathway is frequently activated in T-cell acute lymphoblastic leukemias (T-ALLs). Of the Notch receptors, Notch1 is a recurrent target of gain-of-function mutations and Notch3 is expressed in all T-ALLs, but it is currently unclear how these receptors contribute to T-cell transformation in vivo. We investigated the role of Notch1 and Notch3 in T-ALL progression by a genetic approach, in mice bearing a knockdown mutation in the Ikaros gene that spontaneously develop Notch-dependent T-ALL. While deletion of Notch3 has little effect, T cell-specific deletion of floxed Notch1 promoter/exon 1 sequences significantly accelerates leukemogenesis. Notch1-deleted tumors lack surface Notch1 but express γ-secretase-cleaved intracellular Notch1 proteins. In addition, these tumors accumulate high levels of truncated Notch1 transcripts that are caused by aberrant transcription from cryptic initiation sites in the 3' part of the gene. Deletion of the floxed sequences directly reprograms the Notch1 locus to begin transcription from these 3' promoters and is accompanied by an epigenetic reorganization of the Notch1 locus that is consistent with transcriptional activation. Further, spontaneous deletion of 5' Notch1 sequences occurs in approximately 75% of Ikaros-deficient T-ALLs. These results reveal a novel mechanism for the oncogenic activation of the Notch1 gene after deletion of its main promoter.


Subject(s)
Ikaros Transcription Factor/physiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Promoter Regions, Genetic/genetics , Receptor, Notch1/genetics , Transcriptional Activation/physiology , Animals , Blotting, Northern , Blotting, Western , Cell Transformation, Neoplastic , DNA Primers/chemistry , DNA Primers/genetics , Flow Cytometry , Gene Expression Regulation, Neoplastic , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Mice , Mice, Knockout , Mutation/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA, Messenger/genetics , Receptor, Notch3 , Receptors, Notch/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL