Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Development ; 149(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-35103284

ABSTRACT

The contractile phenotype of smooth muscle cells (SMCs) is transcriptionally controlled by a complex of the DNA-binding protein SRF and the transcriptional co-activator MYOCD. The pathways that activate expression of Myocd and of SMC structural genes in mesenchymal progenitors are diverse, reflecting different intrinsic and extrinsic signaling inputs. Taking the ureter as a model, we analyzed whether Notch signaling, a pathway previously implicated in vascular SMC development, also affects visceral SMC differentiation. We show that mice with a conditional deletion of the unique Notch mediator RBPJ in the undifferentiated ureteric mesenchyme exhibit altered ureter peristalsis with a delayed onset, and decreased contraction frequency and intensity at fetal stages. They also develop hydroureter 2 weeks after birth. Notch signaling is required for precise temporal activation of Myocd expression and, independently, for expression of a group of late SMC structural genes. Based on additional expression analyses, we suggest that a mesenchymal JAG1-NOTCH2/NOTCH3 module regulates visceral SMC differentiation in the ureter in a biphasic and bimodal manner, and that its molecular function differs from that in the vascular system.


Subject(s)
Cell Differentiation , Myocytes, Smooth Muscle/metabolism , Signal Transduction , Ureter/metabolism , Actins/genetics , Actins/metabolism , Animals , Cell Differentiation/drug effects , Diamines/pharmacology , Female , Gene Expression Regulation, Developmental , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Male , Mice , Mice, Knockout , Myocytes, Smooth Muscle/cytology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Thiazoles/pharmacology , Trans-Activators/genetics , Trans-Activators/metabolism , Ureter/cytology , Ureter/growth & development , Viscera/cytology , Viscera/metabolism
2.
Development ; 147(12)2020 06 22.
Article in English | MEDLINE | ID: mdl-32467237

ABSTRACT

Thymus function depends on the epithelial compartment of the thymic stroma. Cortical thymic epithelial cells (cTECs) regulate T cell lineage commitment and positive selection, while medullary (m) TECs impose central tolerance on the T cell repertoire. During thymus organogenesis, these functionally distinct sub-lineages are thought to arise from a common thymic epithelial progenitor cell (TEPC). However, the mechanisms controlling cTEC and mTEC production from the common TEPC are not understood. Here, we show that emergence of the earliest mTEC lineage-restricted progenitors requires active NOTCH signaling in progenitor TEC and that, once specified, further mTEC development is NOTCH independent. In addition, we demonstrate that persistent NOTCH activity favors maintenance of undifferentiated TEPCs at the expense of cTEC differentiation. Finally, we uncover a cross-regulatory relationship between NOTCH and FOXN1, a master regulator of TEC differentiation. These data establish NOTCH as a potent regulator of TEPC and mTEC fate during fetal thymus development, and are thus of high relevance to strategies aimed at generating/regenerating functional thymic tissue in vitro and in vivo.


Subject(s)
Embryonic Development/genetics , Receptors, Notch/metabolism , Thymus Gland/metabolism , Animals , Cell Differentiation , Cell Lineage , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gain of Function Mutation , Gene Expression Regulation, Developmental , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Organogenesis , Receptors, Notch/genetics , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Thymus Gland/cytology , Thymus Gland/growth & development
3.
Nat Commun ; 10(1): 2817, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31249304

ABSTRACT

Sufficient vascular supply is indispensable for brain development and function, whereas dysfunctional blood vessels are associated with human diseases such as vascular malformations, stroke or neurodegeneration. Pericytes are capillary-associated mesenchymal cells that limit vascular permeability and protect the brain by preserving blood-brain barrier integrity. Loss of pericytes has been linked to neurodegenerative changes in genetically modified mice. Here, we report that postnatal inactivation of the Rbpj gene, encoding the transcription factor RBPJ, leads to alteration of cell identity markers in brain pericytes, increases local TGFß signalling, and triggers profound changes in endothelial behaviour. These changes, which are not mimicked by pericyte ablation, imperil vascular stability and induce the acquisition of pathological landmarks associated with cerebral cavernous malformations. In adult mice, loss of Rbpj results in bigger stroke lesions upon ischemic insult. We propose that brain pericytes can acquire deleterious properties that actively enhance vascular lesion formation and promote pathogenic processes.


Subject(s)
Brain/metabolism , Hemangioma, Cavernous, Central Nervous System/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Pericytes/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/cytology , Disease Progression , Female , Hemangioma, Cavernous, Central Nervous System/genetics , Hemangioma, Cavernous, Central Nervous System/pathology , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Male , Mice, Knockout
4.
Reprod Sci ; 26(6): 774-784, 2019 06.
Article in English | MEDLINE | ID: mdl-30213224

ABSTRACT

During decidualization, endometrial stromal cells differentiate into a secretory phenotype to modulate the uterine microenvironment and promote embryo implantation. This highly metabolic process relies on ovarian steroid receptors and glucose transporters. Canonical Notch signaling is mediated by the transcription factor Recombination Signal Binding Protein for Immunoglobulin Kappa J Region (RBPJ). Loss of RBPJ in the mouse uterus (Pgrcre/+Rbpjflox/flox; Rbpj c-KO) results in subfertility in part due to an abnormal uterine-embryonic axis during implantation and, as described herein, decidualization failure. Induced in vivo decidualization in Rbpj c-KO mice was impaired with the downregulation of decidual markers and decreased progesterone receptor (Pgr) signaling. Consistent with in vivo mouse data, RBPJ knockdown during in vitro Human uterine fibroblast (HuF) cell decidualization results in the reduced expression of decidual marker genes along with PGR. Expression of the glucose transporter, SLC2A1, was decreased in the RBPJ-silenced HuF cells, which corresponded to decreased Slc2a1 in the secondary decidual zone of Rbpj c-KO mouse uteri. Exogenous administration of pyruvate, which bypasses the need for glucose, rescues PRL expression in RBPJ-deficient HuF cells. In summary, Notch signaling through RBPJ controls both ovarian steroid receptor PGR and glucose transporter SLC2A1 expression during decidualization, and this dysregulation likely contributes to embryo implantation failure.


Subject(s)
Decidua/physiology , Gene Expression Regulation/physiology , Glucose Transporter Type 1/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Receptors, Progesterone/genetics , Animals , Cell Line , Decidua/chemistry , Female , Fibroblasts , Glucose Transporter Type 1/analysis , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, Knockout , Pyruvic Acid/pharmacology , RNA, Small Interfering/genetics , Receptors, Progesterone/metabolism , Signal Transduction/physiology , Uterus/cytology
5.
PLoS One ; 13(6): e0198609, 2018.
Article in English | MEDLINE | ID: mdl-29889863

ABSTRACT

Macrophages exhibit diverse effector phenotypes depending on the stimuli and their microenvironment. Classically activated macrophages are primed with interferon (IFN)γ and stimulated with pathogen-associated molecular patterns. They produce inflammatory mediators and inflammatory cytokines, such as IL-12. In the presence of immune complexes (ICs), activated macrophages have decreased IL-12 production and increased IL-10 production and presumably act as regulatory macrophages. Notch signaling has been shown to regulate the effector functions of classically activated macrophages. In this study, we investigated whether Notch signaling is active in lipopolysaccharide (LPS)-stimulated macrophages in the presence of ICs. LPS/IC stimulation increased the level of cleaved Notch1 in murine macrophages, while IC stimulation alone did not. Delta-like 4, but not Jagged1, was responsible for generating cleaved Notch1. The activation of Notch signaling by LPS/ICs depended upon NF-κB and MEK/Erk pathway activation. Macrophages with the targeted deletion of Rbpj, which encodes a DNA-binding protein central to canonical Notch signaling, produced significantly less IL-10 upon LPS/IC stimulation. A similar impact on IL-10 production was observed when Notch signaling was inhibited with a gamma-secretase inhibitor (GSI). Defects in NF-κB p50 nuclear localization were observed in GSI-treated macrophages and in Rbpj-/- macrophages, suggesting cross-regulation between the Notch and NF-κB pathways. Transcriptomic analysis revealed that Notch signaling regulates the transcription of genes involved in the cell cycle, macrophage activation, leukocyte migration and cytokine production in LPS/IC-stimulated macrophages. Taken together, these results suggest that the Notch signaling pathway plays an important role in regulating the functions of macrophages activated by LPS and ICs.


Subject(s)
Lipopolysaccharides/pharmacology , Macrophages/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Animals , Antigen-Antibody Complex/pharmacology , Bone Marrow Cells/cytology , Cells, Cultured , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Interferon-gamma/pharmacology , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-12/genetics , Interleukin-12/metabolism , Macrophages/cytology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism
6.
Sci Rep ; 7(1): 13690, 2017 10 20.
Article in English | MEDLINE | ID: mdl-29057904

ABSTRACT

Transgenic mice expressing the Notch-4 intracellular domain (designated Int3) in the mammary gland have two phenotypes exhibited with 100% penetrance: arrest of mammary alveolar/lobular development and mammary tumorigenesis. Notch-4 signaling is mediated primarily through the interaction of Int3 with the transcription repressor/activator Rbpj. Interestingly, WAP-Int3/Rbpj knockout mice have normal mammary gland development but still developed mammary tumors with a slightly longer latency than the WAP-Int3 mice. Thus, Notch-induced mammary tumor development is Rbpj-independent. Here, we show that Int3 activates NF-κB in HC11 cells in absence of Rbpj through an association with the IKK signalosome. Int3 induced the canonical NF-κB activity and P50 phosphorylation in HC11 cells without altering the NF-κB2 pathway. The minimal domain within the Int3 protein required to activate NF-κB consists of the CDC10/Ankyrin (ANK) repeats domain. Treatment of WAP-Int3 tumor bearing mice with an IKK inhibitor resulted in tumor regression. In a soft agar assay, treatment of HC11-Int3 cells with P50-siRNA caused a significant decrease in colony formation. In addition, Wap-Int3/P50 knockout mice did not develop mammary tumors. This data indicates that the activation of NF-κB canonical signaling by Notch-4/Int3 is ANK repeats dependent, Rbpj-independent, and is mediated by IKK activation and P50 phosphorylation causing mammary tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/metabolism , NF-kappa B/metabolism , Receptor, Notch4/metabolism , Animals , Ankyrin Repeat , Cell Line , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice, Knockout , Receptor, Notch4/genetics , Signal Transduction
7.
Sci Rep ; 6: 26874, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27240887

ABSTRACT

Notch signaling is known to control early pancreatic differentiation through Ngn3 repression. In later stages, downstream of Notch, the Presenilins are still required to maintain the endocrine fate allocation. Amongst their multiple targets, it remains unclear which one actually controls the maintenance of the fate of the early islets. Conditional deletions of the Notch effector RBP-Jκ with lineage tracing in Presenilin-deficient endocrine progenitors, demonstrated that this factor is central to the control of the fate through a non-canonical Notch mechanism. RBP-Jκ mice exhibit normal islet morphogenesis and function, however, a fraction of the progenitors fails to differentiate and develop into disorganized masses resembling acinar to ductal metaplasia and chronic pancreatitis. A subsequent deletion of RBP-Jκ in forming ß-cells led to the transdifferentiation into the other endocrine cells types, indicating that this factor still mediates the maintenance of the fate within the endocrine lineage itself. These results highlight the dual importance of Notch signaling for the endocrine lineage. Even after Ngn3 expression, Notch activity is required to maintain both fate and maturation of the Ngn3 progenitors. In a subset of the cells, these alterations of Notch signaling halt their differentiation and leads to acinar to ductal metaplasia.


Subject(s)
Enteroendocrine Cells/metabolism , Gene Expression Regulation, Developmental , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Islets of Langerhans/metabolism , Morphogenesis/genetics , Mouse Embryonic Stem Cells/metabolism , Acinar Cells/cytology , Acinar Cells/metabolism , Animals , Animals, Genetically Modified , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Lineage/genetics , Embryo, Mammalian , Enteroendocrine Cells/cytology , Female , Genes, Reporter , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Integrases/genetics , Integrases/metabolism , Islets of Langerhans/cytology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Metaplasia/genetics , Metaplasia/metabolism , Metaplasia/pathology , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/cytology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Presenilin-1/deficiency , Presenilin-1/genetics , Presenilin-2/deficiency , Presenilin-2/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction
8.
Sci Rep ; 6: 26003, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27188577

ABSTRACT

Physical and chemical insult-induced bone marrow (BM) damage often leads to lethality resulting from the depletion of hematopoietic stem and progenitor cells (HSPCs) and/or a deteriorated BM stroma. Notch signaling plays an important role in hematopoiesis, but whether it is involved in BM damage remains unclear. In this study, we found that conditional disruption of RBP-J, the transcription factor of canonical Notch signaling, increased irradiation sensitivity in mice. Activation of Notch signaling with the endothelial cell (EC)-targeted soluble Dll1 Notch ligand mD1R promoted BM recovery after irradiation. mD1R treatment resulted in a significant increase in myeloid progenitors and monocytes in the BM, spleen and peripheral blood after irradiation. mD1R also enhanced hematopoiesis in mice treated with cyclophosphamide, a chemotherapeutic drug that induces BM suppression. Mechanistically, mD1R increased the proliferation and reduced the apoptosis of myeloid cells in the BM after irradiation. The ß chain cytokine receptor Csf2rb2 was identified as a downstream molecule of Notch signaling in hematopoietic cells. mD1R improved hematopoietic recovery through up-regulation of the hematopoietic expression of Csf2rb2. Our findings reveal the role of Notch signaling in irradiation- and drug-induced BM suppression and establish a new potential therapy of BM- and myelo-suppression induced by radiotherapy and chemotherapy.


Subject(s)
Bone Marrow/physiology , Genetic Predisposition to Disease , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/metabolism , Radiation Injuries, Experimental/physiopathology , Receptors, Interleukin-3/metabolism , Animals , Blood Cells , Bone Marrow/radiation effects , Calcium-Binding Proteins , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Male , Mice, Inbred C57BL , Myeloid Progenitor Cells/physiology , Regeneration , Signal Transduction , Spleen/cytology , Up-Regulation
9.
Proc Natl Acad Sci U S A ; 113(8): 2300-5, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26858409

ABSTRACT

In mammalian reproduction, implantation is one of the most critical events. Failure of implantation and the subsequent decidualization contribute to more than 75% of pregnancy losses in women. Our laboratory has previously reported that inhibition of Notch signaling results in impaired decidualization in both women and a transgenic mouse model. In this study, we generated a Notch gain-of-function transgenic mouse by conditionally overexpressing the Notch1 intracellular domain (N1ICD) in the reproductive tract driven by a progesterone receptor (Pgr) -Cre. We show that the overexpression of N1ICD in the uterus results in complete infertility as a consequence of multiple developmental and physiological defects, including the absence of uterine glands and dysregulation of progesterone and estrogen signaling by a Recombination Signal Binding Protein Jκ-dependent signaling mechanism. We further show that the inhibition of progesterone signaling is caused by hypermethylation of its receptor Pgr by Notch1 overexpression through the transcription factor PU.1 and DNA methyltransferase 3b (Dnmt3b). We have generated a mouse model to study the consequence of increased Notch signaling in female reproduction and provide the first evidence, to our knowledge, that Notch signaling can regulate epigenetic modification of the Pgr.


Subject(s)
Infertility, Female/etiology , Infertility, Female/metabolism , Receptor, Notch1/metabolism , Receptors, Progesterone/metabolism , Uterus/metabolism , Animals , DNA Methylation , Disease Models, Animal , Epigenesis, Genetic , Estradiol/metabolism , Female , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Infertility, Female/genetics , Mice , Mice, Knockout , Mice, Transgenic , Pregnancy , Progesterone/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Notch1/chemistry , Receptor, Notch1/genetics , Receptors, Progesterone/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Up-Regulation , Uterus/pathology
10.
PLoS One ; 10(8): e0135971, 2015.
Article in English | MEDLINE | ID: mdl-26285013

ABSTRACT

Recently we have demonstrated the importance of RBPjk-dependent Notch signaling in the regulation of mesenchymal stem cell (MSC) differentiation during skeletogenesis both in vivo and in vitro. Here we further performed RBPJK loss-of-function experiments to demonstrate for the first time that RBPJK deficient MSC shows enhanced differentiation and osteogenesis acts via up-regulation of the BMP signaling. In the present study, we first compared the spontaneous and osteogenic differentiation in normal and recombination signal binding protein for immunoglobulin kappa J region (RBPJK) deficient human bone marrow-derived mesenchymal stem cells (MSCs). It was found that RBPJK highly expressed in fresh isolated MSCs and its expression was progressing down-regulated during spontaneous differentiation and even greater in osteogenic media inducted differentiation. Deletion of RBPJK in MSCs not only enhances cell spontaneous differentiation, but also significantly accelerates condition media inducted osteogenic differentiation by showing enhanced alkaline phosphatase (ALP) activity, Alizarin red staining, gene expression of Runx2, Osteopontin (OPN), Type I collagen (COL1a1) in culture. Additionally, BMP signaling responsive reporter activity and phosphor-smad1/5/8 expression were also significantly increased upon removal of RBPJK in MSCs. These data proved that inhibition of Notch signaling in MSCs promotes cell osteogenic differentiation by up-regulation of BMP signaling, and RBPJK deficient MSC maybe a better cell population for cell-based bone tissue engineering.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Gene Expression Regulation , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/physiology , Aged , Apoptosis , Blotting, Western , Bone Morphogenetic Protein 1/genetics , Cell Differentiation , Cell Proliferation , Cells, Cultured , Female , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Male , Middle Aged , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
11.
Leukemia ; 29(3): 576-85, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25234168

ABSTRACT

In leukemogenesis, Notch signaling can be up and downregulated in a context-dependent manner. The transcription factor hairy and enhancer of split-1 (Hes1) is well-characterized as a downstream target of Notch signaling. Hes1 encodes a basic helix-loop-helix-type protein, and represses target gene expression. Here, we report that deletion of the Hes1 gene in mice promotes acute myeloid leukemia (AML) development induced by the MLL-AF9 fusion protein. We then found that Hes1 directly bound to the promoter region of the FMS-like tyrosine kinase 3 (FLT3) gene and downregulated the promoter activity. FLT3 was consequently upregulated in MLL-AF9-expressing immortalized and leukemia cells with a Hes1- or RBPJ-null background. MLL-AF9-expressing Hes1-null AML cells showed enhanced proliferation and ERK phosphorylation following FLT3 ligand stimulation. FLT3 inhibition efficiently abrogated proliferation of MLL-AF9-induced Hes1-null AML cells. Furthermore, an agonistic anti-Notch2 antibody induced apoptosis of MLL-AF9-induced AML cells in a Hes1-wild type but not a Hes1-null background. We also accessed two independent databases containing messenger RNA (mRNA) expression profiles and found that the expression level of FLT3 mRNA was negatively correlated with those of HES1 in patient AML samples. These observations demonstrate that Hes1 mediates tumor suppressive roles of Notch signaling in AML development, probably by downregulating FLT3 expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation, Leukemic , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/genetics , fms-Like Tyrosine Kinase 3/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Cell Proliferation , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Transgenic , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Survival Analysis , Transcription Factor HES-1 , fms-Like Tyrosine Kinase 3/metabolism
12.
Mol Neurobiol ; 52(3): 1378-1390, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25344316

ABSTRACT

The outcome of spinal cord injury (SCI) is determined by both neural cell-intrinsic survival pathways and tissue microenvironment-derived signals. Macrophages dominating the inflammatory responses in SCI possess both destructive and reparative potentials, according to their activation status. Notch signaling is involved in both cell survival and macrophage-mediated inflammation, but a comprehensive role of Notch signaling in SCI has been elusive. In this study, we compared the effects of general Notch blockade by a pharmaceutical γ-secretase inhibitor (GSI) and myeloid-specific Notch signal disruption by recombination signal binding protein Jκ (RBP-J) knockout on SCI. The administration of Notch signal inhibitor GSI resulted in worsened hind limb locomotion and exacerbated inflammation. However, mice lacking RBP-J, the critical transcription factor mediating signals from all four mammalian Notch receptors, in myeloid lineage displayed promoted functional recovery, attenuated glial scar formation, improved neuronal survival and axon regrowth, and mitigated inflammatory response after SCI. These benefits were accompanied by enhanced AKT activation in the lesion area after SCI. These findings demonstrate that abrogating Notch signal in myeloid cells ameliorates inflammation response post-SCI and promotes functional recovery, but general pharmaceutical Notch interception has opposite effects. Therefore, clinical intervention of Notch signaling in SCI needs to pinpoint myeloid lineage to avoid the counteractive effects of global inhibition.


Subject(s)
Genetic Therapy , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Inflammation/prevention & control , Myeloid Cells/pathology , Nerve Tissue Proteins/deficiency , Receptors, Notch/antagonists & inhibitors , Signal Transduction/drug effects , Spinal Cord Injuries/therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Cellular Microenvironment , Cicatrix/prevention & control , Gene Expression Regulation/drug effects , Gliosis/prevention & control , Hindlimb/physiopathology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/physiology , Inflammation/physiopathology , Locomotion , Macrophage Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/metabolism , Nerve Regeneration , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Oligopeptides/therapeutic use , Oligopeptides/toxicity , Paraplegia/etiology , Paraplegia/physiopathology , Paraplegia/prevention & control , Recovery of Function , Signal Transduction/physiology , Specific Pathogen-Free Organisms
13.
Development ; 142(1): 41-50, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25480918

ABSTRACT

Genetic data indicate that abrogation of Notch-Rbpj or Wnt-ß-catenin pathways results in the loss of the intestinal stem cells (ISCs). However, whether the effect of Notch is direct or due to the aberrant differentiation of the transit-amplifying cells into post-mitotic goblet cells is unknown. To address this issue, we have generated composite tamoxifen-inducible intestine-specific genetic mouse models and analyzed the expression of intestinal differentiation markers. Importantly, we found that activation of ß-catenin partially rescues the differentiation phenotype of Rbpj deletion mutants, but not the loss of the ISC compartment. Moreover, we identified Bmi1, which is expressed in the ISC and progenitor compartments, as a gene that is co-regulated by Notch and ß-catenin. Loss of Bmi1 resulted in reduced proliferation in the ISC compartment accompanied by p16(INK4a) and p19(ARF) (splice variants of Cdkn2a) accumulation, and increased differentiation to the post-mitotic goblet cell lineage that partially mimics Notch loss-of-function defects. Finally, we provide evidence that Bmi1 contributes to ISC self-renewal.


Subject(s)
Intestines/pathology , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Cell Compartmentation , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p19/genetics , Cyclin-Dependent Kinase Inhibitor p19/metabolism , DNA Repair , Homeostasis , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Intestines/abnormalities , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Polycomb Repressive Complex 1/deficiency , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptors, Notch/deficiency , Transcriptional Activation/genetics , Wnt Proteins/metabolism , beta Catenin/metabolism
14.
Nat Med ; 21(1): 55-61, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25501905

ABSTRACT

CD4+ T cells differentiate into memory T cells that protect the host from subsequent infection. In contrast, autoreactive memory CD4+ T cells harm the body by persisting in the tissues. The underlying pathways controlling the maintenance of memory CD4+ T cells remain undefined. We show here that memory CD4+ T cell survival is impaired in the absence of the Notch signaling protein known as recombination signal binding protein for immunoglobulin κ J region (Rbpj). Treatment of mice with a Notch inhibitor reduced memory CD4+ T cell numbers and prevented the recurrent induction of experimental autoimmune encephalomyelitis. Rbpj-deficient CD4+ memory T cells exhibit reduced glucose uptake due to impaired AKT phosphorylation, resulting in low Glut1 expression. Treating mice with pyruvic acid, which bypasses glucose uptake and supplies the metabolite downstream of glucose uptake, inhibited the decrease of autoimmune memory CD4+ T cells in the absence of Notch signaling, suggesting memory CD4+ T cell survival relies on glucose metabolism. Together, these data define a central role for Notch signaling in maintaining memory CD4+ T cells through the regulation of glucose uptake.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Glucose/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , CD4-Positive T-Lymphocytes/metabolism , Glucose/immunology , Glucose Transporter Type 1/biosynthesis , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Oncogene Protein v-akt/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Notch/antagonists & inhibitors
15.
Arterioscler Thromb Vasc Biol ; 34(12): 2624-31, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25324571

ABSTRACT

OBJECTIVE: The goal of the present study was to identify novel mechanisms that regulate smooth muscle cell (SMC) differentiation marker gene expression. APPROACH AND RESULTS: We demonstrate that the CArG-containing regions of many SMC-specific promoters are imbedded within CpG islands. A previously identified GC repressor element in the SM myosin heavy chain (MHC) promoter was highly methylated in cultured aortic SMC but not in the aorta, and this difference was inversely correlated with SM MHC expression. Using an affinity chromatography/mass spectroscopy-based approach, we identified the multifunctional Notch transcription factor, recombination signal binding protein for immunoglobulin κ J region (RBPJ), as a methylated GC repressor-binding protein. RBPJ protein levels and binding to the endogenous SM MHC GC repressor were enhanced by platelet-derived growth factor-BB treatment. A methylation mimetic mutation to the GC repressor that facilitated RBPJ binding inhibited SM MHC promoter activity as did overexpression of RBPJ. Consistent with this, knockdown of RBPJ in phenotypically modulated human aortic SMC enhanced endogenous SMC marker gene expression, an effect likely mediated by increased recruitment of serum response factor and Pol II to the SMC-specific promoters. In contrast, the depletion of RBPJ in differentiated transforming growth factor-ß-treated SMC inhibited SMC-specific gene activation, supporting the idea that the effects of RBPJ/Notch signaling are context dependent. CONCLUSIONS: Our results indicate that methylation-dependent binding of RBPJ to a GC repressor element can negatively regulate SM MHC promoter activity and that RBPJ can inhibit SMC marker gene expression in phenotypically modulated SMC. These results will have important implications on the regulation of SMC phenotype and on Notch-dependent transcription.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Myosin Heavy Chains/genetics , Promoter Regions, Genetic , Smooth Muscle Myosins/genetics , Animals , Base Sequence , Becaplermin , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , CpG Islands , DNA Methylation , GC Rich Sequence , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, Knockout , Molecular Sequence Data , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Protein Binding , Proto-Oncogene Proteins c-sis/metabolism , Receptors, Notch/metabolism , Signal Transduction
16.
Neuron ; 83(5): 1085-97, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25189209

ABSTRACT

The activity of adult stem cells is regulated by signals emanating from the surrounding tissue. Many niche signals have been identified, but it is unclear how they influence the choice of stem cells to remain quiescent or divide. Here we show that when stem cells of the adult hippocampus receive activating signals, they first induce the expression of the transcription factor Ascl1 and only subsequently exit quiescence. Moreover, lowering Ascl1 expression reduces the proliferation rate of hippocampal stem cells, and inactivating Ascl1 blocks quiescence exit completely, rendering them unresponsive to activating stimuli. Ascl1 promotes the proliferation of hippocampal stem cells by directly regulating the expression of cell-cycle regulatory genes. Ascl1 is similarly required for stem cell activation in the adult subventricular zone. Our results support a model whereby Ascl1 integrates inputs from both stimulatory and inhibitory signals and converts them into a transcriptional program activating adult neural stem cells.


Subject(s)
Adult Stem Cells/cytology , Gene Expression Regulation, Developmental/genetics , Hippocampus/cytology , Neurogenesis/genetics , Adult Stem Cells/metabolism , Age Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation , Cerebral Ventricles/cytology , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 1/metabolism , Gene Expression Regulation, Developmental/drug effects , Glial Fibrillary Acidic Protein/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Kainic Acid/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Box Domain Proteins/metabolism , p300-CBP Transcription Factors/metabolism
17.
Development ; 141(19): 3782-92, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25209249

ABSTRACT

Arteriovenous malformations (AVMs) are tortuous vessels characterized by arteriovenous (AV) shunts, which displace capillaries and shunt blood directly from artery to vein. Notch signaling regulates embryonic AV specification by promoting arterial, as opposed to venous, endothelial cell (EC) fate. To understand the essential role of endothelial Notch signaling in postnatal AV organization, we used inducible Cre-loxP recombination to delete Rbpj, a mediator of canonical Notch signaling, from postnatal ECs in mice. Deletion of endothelial Rbpj from birth resulted in features of AVMs by P14, including abnormal AV shunting and tortuous vessels in the brain, intestine and heart. We further analyzed brain AVMs, as they pose particular health risks. Consistent with AVM pathology, we found cerebral hemorrhage, hypoxia and necrosis, and neurological deficits. AV shunts originated from capillaries (and possibly venules), with the earliest detectable morphological abnormalities in AV connections by P8. Prior to AV shunt formation, alterations in EC gene expression were detected, including decreased Efnb2 and increased Pai1, which encodes a downstream effector of TGFß signaling. After AV shunts had formed, whole-mount immunostaining showed decreased Efnb2 and increased Ephb4 expression within AV shunts, suggesting that ECs were reprogrammed from arterial to venous identity. Deletion of Rbpj from adult ECs led to tortuosities in gastrointestinal, uterine and skin vascular beds, but had mild effects in the brain. Our results demonstrate a temporal requirement for Rbpj in postnatal ECs to maintain proper artery, capillary and vein organization and to prevent abnormal AV shunting and AVM pathogenesis.


Subject(s)
Arteriovenous Malformations/genetics , Arteriovenous Malformations/pathology , Endothelium, Vascular/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Gene Deletion , Gene Expression Profiling , Image Processing, Computer-Assisted , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Microscopy, Fluorescence , Real-Time Polymerase Chain Reaction , Receptor, EphB4/metabolism
18.
Proc Natl Acad Sci U S A ; 111(16): 5986-91, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24711412

ABSTRACT

The gastrointestinal tract comes into direct contact with environmental agents, including bacteria, viruses, and foods. Intestine-specific subsets of immune cells maintain gut homeostasis by continuously sampling luminal antigens and maintaining immune tolerance. CD11c(+)CX3CR1(+) cells sample luminal antigens in the small intestine and contribute to the trafficking of bacteria to lymph nodes under dysbiotic conditions. The molecular mechanisms crucial for the differentiation of CD11c(+)CX3CR1(+) cells remain unclear. Here we demonstrate that the Notch1- or Notch2-Rbpj axis is essential for the development of CD11c(+)CX3CR1(+) cells. In mice in which Rbpj or Notch1 and Notch2 were deleted from CD11c(+) cells, there was a deficit of CD11c(+)CX3CR1(+) cells and an accumulation of CD11c(low)CX3CR1(+) cells. The CD11c(low)CX3CR1(+) cells could not differentiate to CD11c(+)CX3CR1(+) cells, suggesting that CD11c(low)CX3CR1(+) cells represent a lineage distinct from CD11c(+)CX3CR1(+) cells. These data indicate that Notch signaling is essential for lineage fixation of intestinal CD11c(+)CX3CR1(+) cells.


Subject(s)
CD11c Antigen/metabolism , Cell Differentiation , Intestine, Small/cytology , Receptors, Chemokine/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , CX3C Chemokine Receptor 1 , Cell Count , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Mice, Inbred C57BL
19.
Am J Pathol ; 184(5): 1479-88, 2014 May.
Article in English | MEDLINE | ID: mdl-24631193

ABSTRACT

The potential for intrahepatic bile duct (IHBD) regeneration in patients with bile duct insufficiency diseases is poorly understood. Notch signaling and Hnf6 have each been shown to be important for the morphogenesis of IHBDs in mice. One congenital pediatric liver disease characterized by reduced numbers of IHBDs, Alagille syndrome, is associated with mutations in Notch signaling components. Therefore, we investigated whether liver cell plasticity could contribute to IHBD regeneration in mice with disruptions in Notch signaling and Hnf6. We studied a mouse model of bile duct insufficiency with liver epithelial cell-specific deficiencies in Hnf6 and Rbpj, a mediator of canonical Notch signaling. Albumin-Cre Hnf6(flox/flox)Rbpj(flox/flox) mice initially developed no peripheral bile ducts. The evolving postnatal liver phenotype was analyzed using IHBD resin casting, immunostaining, and serum chemistry. With age, Albumin-Cre Hnf6(flox/flox)Rbpj(flox/flox) mice mounted a ductular reaction extending through the hepatic tissue and then regenerated communicating peripheral IHBD branches. Rbpj and Hnf6 were determined to remain absent from biliary epithelial cells constituting the ductular reaction and the regenerated peripheral IHBDs. We report the expression of Sox9, a marker of biliary epithelial cells, in cells expressing hepatocyte markers. Tissue analysis indicates that reactive ductules did not arise directly from preexisting hilar IHBDs. We conclude that liver cell plasticity is competent for regeneration of IHBDs independent of Notch signaling via Rbpj and Hnf6.


Subject(s)
Bile Ducts, Intrahepatic/physiology , Hepatocyte Nuclear Factor 6/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Receptors, Notch/metabolism , Regeneration/physiology , Animals , Epithelial Cells/metabolism , Hepatocyte Nuclear Factor 6/deficiency , Hepatocytes/metabolism , Imaging, Three-Dimensional , Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Immunohistochemistry , Keratin-19/metabolism , Mice, Knockout , Plant Lectins/metabolism , Portal Vein/metabolism , SOX9 Transcription Factor/metabolism
20.
Int J Clin Exp Pathol ; 6(7): 1261-71, 2013.
Article in English | MEDLINE | ID: mdl-23826407

ABSTRACT

Somatosensory ganglia including dorsal root ganglion (DRG) and trigeminal ganglion (TG) are derived from a common pool of neural crest stem cells (NCCs), and are good systems to study the mechanisms of neurogenesis and gliogenesis. Previous studies have reported that deletion of Rbpj, a critical integrator of activation signals from all Notch receptors, in NCCs and their derived cells resulted in the delayed gliogenesis at early stage and a loss of glial cells at later stage in the DRG. But the phenotypes in the TG have not been described. Here we reported although the gliogenesis was also delayed initially in Rbpj-deficient TG, it was recovered as the development progressed, as shown by the presence of large number of glial cells in the TG at later stages. However, neuronal reduction was observed in Rbpj-deficient TG, which is similar to what observed in Rbpj-deficient DRG. Taken together, our data indicate the function of Rbpj is diversified and context dependent in the gliogenesis of somatosensory ganglia.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency , Neuroglia/metabolism , Trigeminal Ganglion/metabolism , Animals , Biomarkers/metabolism , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neurogenesis , Neurons/metabolism , Phenotype , Receptors, Nerve Growth Factor/metabolism , SOXE Transcription Factors/metabolism , Time Factors , Trigeminal Ganglion/embryology
SELECTION OF CITATIONS
SEARCH DETAIL