Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 196
Filter
1.
Protein Sci ; 33(4): e4947, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38511488

ABSTRACT

Notch signaling in humans is mediated by four paralogous receptors that share conserved architectures and possess overlapping, yet non-redundant functions. The receptors share a canonical activation pathway wherein upon extracellular ligand binding, the Notch intracellular domain (NICD) is cleaved from the membrane and translocates to the nucleus where its N-terminal RBP-j-associated molecule (RAM) region and ankyrin repeat (ANK) domain bind transcription factor CSL and recruit co-activator Mastermind-like-1 (MAML1) to activate transcription. However, different paralogs can lead to distinct outcomes. To better understand paralog-specific differences in Notch signaling, we performed a thermodynamic analysis of the Notch transcriptional activation complexes for all four Notch paralogs using isothermal titration calorimetry. Using chimeric constructs, we find that the RAM region is the primary determinant of stability of binary RAMANK:CSL complexes, and that the ANK regions are largely the determinants of MAML1 binding to pre-formed RAMANK:CSL complexes. Free energies of these binding reactions (ΔGRA and ΔGMAML) vary among the four Notch paralogs, although variations for Notch2, 3, and 4 offset in the free energy of the ternary complex (ΔGTC, where ΔGTC = ΔGRA + ΔGMAML). To probe how these affinity differences affect Notch signaling, we performed transcriptional activation assays with the paralogous and chimeric NICDs, and analyzed the results with an independent multiplicative model that quantifies contributions of the paralogous RAM, ANK, and C-terminal regions (CTR) to activation. This analysis shows that transcription activation correlates with ΔGTC, but that activation is further modified by CTR identity in a paralog-specific way.


Subject(s)
Gene Expression Regulation , Receptors, Notch , Humans , Transcriptional Activation , Receptors, Notch/genetics , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Protein Binding , Thermodynamics , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism
2.
Structure ; 31(5): 584-594.e5, 2023 05 04.
Article in English | MEDLINE | ID: mdl-36977409

ABSTRACT

The Notch signaling pathway, an important cell fate determination pathway, is modulated by the ubiquitin ligase Deltex. Here, we investigate the structural basis for Deltex-Notch interaction. We used nuclear magnetic resonance (NMR) spectroscopy to assign the backbone of the Drosophila Deltex WWE2 domain and mapped the binding site of the Notch ankyrin (ANK) domain to the N-terminal WWEA motif. Using cultured Drosophila S2R+ cells, we find that point substitutions within the ANK-binding surface of Deltex disrupt Deltex-mediated enhancement of Notch transcriptional activation and disrupt ANK binding in cells and in vitro. Likewise, ANK substitutions that disrupt Notch-Deltex heterodimer formation in vitro block disrupt Deltex-mediated stimulation of Notch transcription activation and diminish interaction with full-length Deltex in cells. Surprisingly, the Deltex-Notch intracellular domain (NICD) interaction is not disrupted by deletion of the Deltex WWE2 domain, suggesting a secondary Notch-Deltex interaction. These results show the importance of the WWEA:ANK interaction in enhancing Notch signaling.


Subject(s)
Ankyrins , Drosophila Proteins , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Notch/genetics , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Drosophila/metabolism , Magnetic Resonance Spectroscopy
3.
Anal Biochem ; 656: 114881, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36067866

ABSTRACT

The Notch pathway represents evolutionarily conserved intercellular signaling essential for cell-to-cell communication during development. Dysregulation of Notch signaling has been implicated in various diseases, and its control represents a potential cancer treatment strategy. Notch signaling is initiated by the interaction of NOTCH receptors with their ligands on neighboring cells. Therefore, the truncated NOTCH ectodomain, composed mainly of tandem repeats of epidermal growth factor-like (EGF) domains, serves as a decoy molecule that competes for ligand binding and thus inhibits ligand-dependent Notch signaling. Although full-length NOTCH EGF repeats exhibited potent Notch inhibitory activity, they were poorly produced in the transfected cells. This study evaluated the effect of EGF domain-modifying glycosyltransferases on the secretion of NOTCH EGF repeats. Our results in HEK293T cells revealed that, unlike the effect on endogenous NOTCH receptors, overexpressed EGF domain-specific O-GlcNAc transferase (EOGT) markedly enhanced the secretion of NOTCH1 EGF repeats in an enzyme activity-dependent manner. The co-expression of protein O-glucosyltransferase 1 further manifested the effect of EOGT. The resultant changes in O-glycosylation of NOTCH3 were evaluated by label-free glycopeptide quantification. This study provides an experimental strategy to efficiently generate NOTCH EGF repeats by manipulating the expression of glycosyltransferases that alter the O-glycosylation of EGF domains.


Subject(s)
Epidermal Growth Factor , Receptors, Notch , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/metabolism , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Glycopeptides , Glycosylation , HEK293 Cells , Humans , Ligands , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Tandem Repeat Sequences
4.
Cell Commun Signal ; 20(1): 66, 2022 05 18.
Article in English | MEDLINE | ID: mdl-35585601

ABSTRACT

Notch signalling pathway plays a key role in metazoan biology by contributing to resolution of binary decisions in the life cycle of cells during development. Outcomes such as proliferation/differentiation dichotomy are resolved by transcriptional remodelling that follows a switch from Notchon to Notchoff state, characterised by dissociation of Notch intracellular domain (NICD) from DNA-bound RBPJ. Here we provide evidence that transitioning to the Notchoff state is regulated by heat flux, a phenomenon that aligns resolution of fate dichotomies to mitochondrial activity. A combination of phylogenetic analysis and computational biochemistry was utilised to disclose structural adaptations of Notch1 ankyrin domain that enabled function as a sensor of heat flux. We then employed DNA-based micro-thermography to measure heat flux during brain development, followed by analysis in vitro of the temperature-dependent behaviour of Notch1 in mouse neural progenitor cells. The structural capacity of NICD to operate as a thermodynamic sensor in metazoans stems from characteristic enrichment of charged acidic amino acids in ß-hairpins of the ankyrin domain that amplify destabilising inter-residue electrostatic interactions and render the domain thermolabile. The instability emerges upon mitochondrial activity which raises the perinuclear and nuclear temperatures to 50 °C and 39 °C, respectively, leading to destabilization of Notch1 transcriptional complex and transitioning to the Notchoff state. Notch1 functions a metazoan thermodynamic sensor that is switched on by intercellular contacts, inputs heat flux as a proxy for mitochondrial activity in the Notchon state via the ankyrin domain and is eventually switched off in a temperature-dependent manner. Video abstract.


Subject(s)
Ankyrins , Neural Stem Cells , Receptors, Notch , Animals , Ankyrins/chemistry , Ankyrins/metabolism , Mice , Neural Stem Cells/chemistry , Neural Stem Cells/metabolism , Phylogeny , Protein Domains , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Signal Transduction , Thermodynamics
5.
Cell Death Dis ; 12(11): 966, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34667161

ABSTRACT

Numb, a stem cell fate determinant, acts as a tumor suppressor and is closely related to a wide variety of malignancies. Intrahepatic cholangiocarcinoma (iCCA) originates from hepatic progenitors (HPCs); however, the role of Numb in HPC malignant transformation and iCCA development is still unclear. A retrospective cohort study indicated that Numb was frequently decreased in tumor tissues and suggests poor prognosis in iCCA patients. Consistently, in a chemically induced iCCA mouse model, Numb was downregulated in tumor cells compared to normal cholangiocytes. In diet-induced chronic liver injury mouse models, Numb ablation significantly promoted histological impairment, HPC expansion, and tumorigenesis. Similarly, Numb silencing in cultured iCCA cells enhanced cell spheroid growth, invasion, metastasis, and the expression of stem cell markers. Mechanistically, Numb was found to bind to the Notch intracellular domain (NICD), and Numb ablation promoted Notch signaling; this effect was reversed when Notch signaling was blocked by γ-secretase inhibitor treatment. Our results suggested that loss of Numb plays an important role in promoting HPC expansion, HPC malignant transformation, and, ultimately, iCCA development in chronically injured livers. Therapies targeting suppressed Numb are promising for the treatment of iCCA.


Subject(s)
Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , Liver/pathology , Membrane Proteins/deficiency , Nerve Tissue Proteins/deficiency , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/metabolism , Animals , Bile Duct Neoplasms/genetics , Body Weight , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Proliferation , Cholangiocarcinoma/genetics , Down-Regulation/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Ki-67 Antigen/metabolism , Liver Cirrhosis/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organ Size , Prognosis , Protein Domains , Receptors, Notch/chemistry , Transcription Factor HES-1/metabolism
6.
Cell Rep ; 34(13): 108905, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33789105

ABSTRACT

Ogt catalyzed O-linked N-acetylglucosamine (O-GlcNAcylation, O-GlcNAc) plays an important function in diverse biological processes and diseases. However, the roles of Ogt in regulating neurogenesis remain largely unknown. Here, we show that Ogt deficiency or depletion in adult neural stem/progenitor cells (aNSPCs) leads to the diminishment of the aNSPC pool and aberrant neurogenesis and consequently impairs cognitive function in adult mice. RNA sequencing reveals that Ogt deficiency alters the transcription of genes relating to cell cycle, neurogenesis, and neuronal development. Mechanistic studies show that Ogt directly interacts with Notch1 and catalyzes the O-GlcNAc modification of Notch TM/ICD fragment. Decreased O-GlcNAc modification of TM/ICD increases the binding of E3 ubiquitin ligase Itch to TM/ICD and promotes its degradation. Itch knockdown rescues neurogenic defects induced by Ogt deficiency in vitro and in vivo. Our findings reveal the essential roles and mechanisms of Ogt and O-GlcNAc modification in regulating mammalian neurogenesis and cognition.


Subject(s)
Aging/metabolism , N-Acetylglucosaminyltransferases/metabolism , Neurogenesis , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/enzymology , Acetylglucosamine/metabolism , Animals , Biocatalysis , Cell Differentiation , Cell Proliferation , Gene Deletion , Glycosylation , HEK293 Cells , Humans , Memory , Mice, Transgenic , N-Acetylglucosaminyltransferases/deficiency , Proteolysis , Receptors, Notch/chemistry , Stem Cells/cytology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
7.
Annu Rev Biophys ; 50: 157-189, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33534608

ABSTRACT

Notch signaling is a conserved system of communication between adjacent cells, influencing numerous cell fate decisions in the development of multicellular organisms. Aberrant signaling is also implicated in many human pathologies. At its core, Notch has a mechanotransduction module that decodes receptor-ligand engagement at the cell surface under force to permit proteolytic cleavage of the receptor, leading to the release of the Notch intracellular domain (NICD). NICD enters the nucleus and acts as a transcriptional effector to regulate expression of Notch-responsive genes. In this article, we review and integrate current understanding of the detailed molecular basis for Notch signal transduction, highlighting quantitative, structural, and dynamic features of this developmentally central signaling mechanism. We discuss the implications of this mechanistic understanding for the functionality of the signaling pathway in different molecular and cellular contexts.


Subject(s)
Receptors, Notch/metabolism , Signal Transduction , Animals , Biophysical Phenomena , Cell Nucleus/metabolism , Humans , Receptors, Notch/chemistry , Receptors, Notch/genetics
8.
Oxid Med Cell Longev ; 2020: 1809408, 2020.
Article in English | MEDLINE | ID: mdl-33149805

ABSTRACT

Notch receptors are transmembrane proteins that are members of the epidermal growth factor-like family. These receptors are widely expressed on the cell surface and are highly conserved. Binding to ligands on adjacent cells results in cleavage of these receptors, and their intracellular domains translocate into the nucleus, where target gene transcription is initiated. In the mammalian kidney, Notch receptors are activated during nephrogenesis and become silenced in the normal kidney after birth. Reactivation of Notch signaling in the adult kidney could be due to the genetic activation of Notch signaling or kidney injury. Notch3 is a mammalian heterodimeric transmembrane receptor in the Notch gene family. Notch3 activation is significantly increased in various glomerular diseases, renal tubulointerstitial diseases, glomerular sclerosis, and renal fibrosis and mediates disease occurrence and development. Here, we discuss numerous recently published papers describing the role of Notch3 signaling in kidney disease.


Subject(s)
Kidney Diseases/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Humans , Kidney/metabolism , Kidney/pathology , Ligands , Models, Biological , Receptors, Notch/chemistry
9.
PLoS One ; 15(11): e0234101, 2020.
Article in English | MEDLINE | ID: mdl-33232323

ABSTRACT

Notch signaling is essential for multicellular life, regulating core functions such as cellular identity, differentiation, and fate. These processes require highly sensitive systems to avoid going awry, and one such regulatory mechanism is through Notch intracellular domain dimerization. Select Notch target genes contain sequence-paired sites (SPS); motifs in which two Notch transcriptional activation complexes can bind and interact through Notch's ankyrin domain, resulting in enhanced transcriptional activation. This mechanism has been mostly studied through Notch1, and to date, the abilities of the other Notch family members have been left unexplored. Through the utilization of minimalized, SPS-driven luciferase assays, we were able to test the functional capacity of Notch dimers. Here we show that the Notch 2 and 3 NICDs also exhibit dimerization-induced signaling, following the same stringent requirements as seen with Notch1. Furthermore, our data suggested that Notch4 may also exhibit dimerization-induced signaling, although the amino acids required for Notch4 NICD dimerization appear to be different than those required for Notch 1, 2, and 3 NICD dimerization. Interestingly, we identified a mechanical difference between canonical and cryptic SPSs, leading to differences in their dimerization-induced regulation. Finally, we profiled the Notch family members' SPS gap distance preferences and found that they all prefer a 16-nucleotide gap, with little room for variation. In summary, this work highlights the potent and highly specific nature of Notch dimerization and refines the scope of this regulatory function.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Mutagenesis , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Animals , Base Sequence , HEK293 Cells , Humans , Mice , Promoter Regions, Genetic , Protein Binding , Protein Domains , Protein Multimerization , Receptor, Notch2/chemistry , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Receptor, Notch3/chemistry , Receptor, Notch3/genetics , Receptor, Notch3/metabolism , Receptor, Notch4/chemistry , Receptor, Notch4/genetics , Receptor, Notch4/metabolism , Receptors, Notch/genetics , Signal Transduction , Transcriptional Activation
10.
Nat Commun ; 11(1): 2163, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32358530

ABSTRACT

Angiogenesis is a necessary process for solid tumor growth. Cellular markers for endothelial cell proliferation are potential targets for identifying the vasculature of tumors in homeostasis. Here we customize the behaviors of engineered cells to recognize Apj, a surface marker of the neovascular endothelium, using synthetic Notch (synNotch) receptors. We designed apelin-based synNotch receptors (AsNRs) that can specifically interact with Apj and then stimulate synNotch pathways. Cells engineered with AsNRs have the ability to sense the proliferation of endothelial cells (ECs). Designed for different synNotch pathways, engineered cells express different proteins to respond to angiogenic signals; therefore, angiogenesis can be detected by cells engineered with AsNRs. Furthermore, T cells customized with AsNRs can sense the proliferation of vascular endothelial cells. As solid tumors generally require vascular support, AsNRs are potential tools for the detection and therapy of a variety of solid tumors in adults.


Subject(s)
Apelin/chemistry , Apelin/metabolism , Neovascularization, Pathologic/metabolism , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Animals , Apelin Receptors/metabolism , Blotting, Western , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/physiology , Endothelial Cells/metabolism , Flow Cytometry , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Immunotherapy , Male , Mice , Mice, Inbred C57BL
11.
Commun Biol ; 3(1): 116, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32170210

ABSTRACT

Notch signaling is highly conserved in most animals and plays critical roles during neurogenesis as well as embryonic development. Synthetic Notch-based systems, modeled from Notch receptors, have been developed to sense and respond to a specific extracellular signal. Recent advancement of synNotch has shown promise for future use in cellular engineering to treat cancers. However, synNotch from Morsut et al. (2016) has a high level of ligand-independent activation, which limits its application. Here we show that adding an intracellular hydrophobic sequence (QHGQLWF, named as RAM7) present in native Notch, significantly reduced ligand-independent activation. Our enhanced synthetic Notch receptor (esNotch) demonstrates up to a 14.6-fold reduction in ligand-independent activation, without affecting its antigen-induced activation efficiency. Our work improves a previously reported transmembrane receptor and provides a powerful tool to develop better transmembrane signaling transduction modules for further advancement of eukaryotic synthetic biology.


Subject(s)
Cell Engineering/methods , Receptors, Artificial/chemistry , Receptors, Artificial/metabolism , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Amino Acid Sequence , Antigens/metabolism , Cell Membrane/metabolism , Cloning, Molecular/methods , HEK293 Cells , Humans , Ligands , Plasmids/genetics , Protein Domains , Proteolysis , Receptors, Notch/genetics , Signal Transduction/genetics , Single-Chain Antibodies , Synthetic Biology/methods , Transfection
12.
Biochem Biophys Res Commun ; 526(1): 184-190, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32201074

ABSTRACT

The Notch signaling pathway is highly conserved and essential in animal development and tissue homeostasis. Regulation of Notch signaling is a crucial process for human health. Ligands initiate a signal cascade by binding to Notch receptors expressed on the neighboring cell. Notch receptors interact with ligands through their epidermal growth factor-like repeats (EGF repeats). Most EGF repeats are modified by O-glycosylation with residues, such as O-linked N-acetylglucosamine (O-GlcNAc), O-fucose, and O-glucose. A recent study revealed the distinct roles of these O-glycans in ligand binding, processing, and trafficking of Notch receptors. In particular, O-GlcNAc glycans are essential for Delta-like (DLL) ligand-mediated Notch signaling. In this study, we showed that O-GlcNAc promotes Notch1 trafficking to the cell surfaces under the condition that O-fucose and O-glucose are removed from consecutive EGF repeats of Notch1. Through in vitro experiments, we showed that O-GlcNAc mediates the stability of EGF domains in the same manner as O-fucose and O-glucose. Thus, O-GlcNAc on EGF domains possesses a shared function in the stability of EGF domains and Notch1 trafficking.


Subject(s)
Epidermal Growth Factor/chemistry , Extracellular Space/metabolism , Glucosamine/metabolism , Protein Folding , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Drosophila/metabolism , Fucose/metabolism , Glucose/metabolism , HEK293 Cells , Humans , Mice , Mutant Proteins/metabolism , Mutation/genetics , N-Acetylglucosaminyltransferases/metabolism , Polysaccharides/metabolism , Protein Domains , Protein Stability , Protein Transport
13.
Sci Rep ; 10(1): 1290, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31992775

ABSTRACT

Cancer stem cells (CSCs) have the ability to self-renew and induce drug resistance and recurrence in colorectal cancer (CRC). As current chemotherapy doesn't eliminate CSCs completely, there is a need to identify novel agents to target them. We investigated the effects of cucurbitacin B (C-B) or I (C-I), a natural compound that exists in edible plants (bitter melons, cucumbers, pumpkins and zucchini), against CRC. C-B or C-I inhibited proliferation, clonogenicity, induced G2/M cell-cycle arrest and caspase-mediated-apoptosis of CRC cells. C-B or C-I suppressed colonosphere formation and inhibited expression of CD44, DCLK1 and LGR5. These compounds inhibited notch signaling by reducing the expression of Notch 1-4 receptors, their ligands (Jagged 1-2, DLL1,3,4), γ-secretase complex proteins (Presenilin 1, Nicastrin), and downstream target Hes-1. Molecular docking showed that C-B or C-I binds to the ankyrin domain of Notch receptor, which was confirmed using the cellular thermal shift assay. Finally, C-B or C-I inhibited tumor xenograft growth in nude mice and decreased the expression of CSC-markers and notch signaling proteins in tumor tissues. Together, our study suggests that C-B and C-I inhibit colon cancer growth by inhibiting Notch signaling pathway.


Subject(s)
Colonic Neoplasms/drug therapy , Molecular Docking Simulation , Receptors, Notch , Signal Transduction/drug effects , Triterpenes , Animals , Colonic Neoplasms/chemistry , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HCT116 Cells , Humans , Male , Mice , Mice, Nude , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Protein Domains , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Triterpenes/chemistry , Triterpenes/pharmacology , Xenograft Model Antitumor Assays
14.
Dev Growth Differ ; 62(1): 15-34, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31943162

ABSTRACT

Notch signaling is involved in the development of almost all organ systems and is required post-developmentally to modulate tissue homeostasis. Rare variants in Notch signaling pathway genes are found in patients with rare Mendelian disorders, while unique or recurrent somatic mutations in a similar set of genes are identified in cancer. The human genome contains four genes that encode Notch receptors, NOTCH1-4, all of which are linked to genetic diseases and cancer. Although some mutations have been classified as clear loss- or gain-of-function alleles based on cellular or rodent based assay systems, the functional consequence of many variants/mutations in human Notch receptors remain unknown. In this review, I will first provide an overview of the domain structure of Notch receptors and discuss how each module is known to regulate Notch signaling activity in vivo using the Drosophila Notch receptor as an example. Next, I will introduce some interesting mutant alleles that have been isolated in the fly Notch gene over the past > 100 years of research and discuss how studies of these mutations have facilitated the understanding of Notch biology. By identifying unique alleles of the fly Notch gene through forward genetic screens, mapping their molecular lesions and characterizing their phenotypes in depth, one can begin to unravel new mechanistic insights into how different domains of Notch fine-tune signaling output. Such information can be useful in deciphering the functional consequences of rare variants/mutations in human Notch receptors, which in turn can influence disease management and therapy.


Subject(s)
Drosophila Proteins , Mutation, Missense , Receptors, Notch , Amino Acid Substitution , Animals , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Protein Domains , Receptors, Notch/chemistry , Receptors, Notch/genetics , Receptors, Notch/metabolism , Structure-Activity Relationship
15.
Sci Signal ; 12(606)2019 11 05.
Article in English | MEDLINE | ID: mdl-31690634

ABSTRACT

Canonical Notch signaling relies on regulated proteolysis of the receptor Notch to generate a nuclear effector that induces the transcription of Notch-responsive genes. In higher organisms, one Notch-responsive gene that is activated in many different cell types encodes the Notch-regulated ankyrin repeat protein (NRARP), which acts as a negative feedback regulator of Notch responses. Here, we showed that NRARP inhibited the growth of Notch-dependent T cell acute lymphoblastic leukemia (T-ALL) cell lines and bound directly to the core Notch transcriptional activation complex (NTC), requiring both the transcription factor RBPJ and the Notch intracellular domain (NICD), but not Mastermind-like proteins or DNA. The crystal structure of an NRARP-NICD1-RBPJ-DNA complex, determined to 3.75 Å resolution, revealed that the assembly of NRARP-NICD1-RBPJ complexes relied on simultaneous engagement of RBPJ and NICD1, with the three ankyrin repeats of NRARP extending the Notch1 ankyrin repeat stack. Mutations at the NRARP-NICD1 interface disrupted entry of the proteins into NTCs and abrogated feedback inhibition in Notch signaling assays in cultured cells. Forced expression of NRARP reduced the abundance of NICD in cells, suggesting that NRARP may promote the degradation of NICD. These studies establish the structural basis for NTC engagement by NRARP and provide insights into a critical negative feedback mechanism that regulates Notch signaling.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/metabolism , Neoplasm Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Notch/metabolism , Signal Transduction , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Jurkat Cells , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Mutation , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Structure, Quaternary , Receptors, Notch/chemistry , Receptors, Notch/genetics
16.
Int J Mol Med ; 44(4): 1574-1584, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31364722

ABSTRACT

The Notch receptor serves a fundamental role in the regulation of cell fate determination through intracellular signal transmission. Mutation of the Notch receptor results in abnormal active signaling, leading to the development of diseases involving abnormal cell growth, including malignant tumors. Therefore, the Notch signaling pathway is a useful pharmacological target for the treatment of cancer. In the present study, a compound screening system was designed to identify inhibitors of the Notch signaling targeting Notch intracellular domain (NICD). A total of 9,600 compounds were analyzed using the Michigan Cancer Foundation­7 (MCF7) human breast adenocarcinoma cell line and the SH­SY5Y human neuroblastoma cell line with the reporter assay system using an artificial protein encoding a partial Notch carboxyl­terminal fragment fused to the Gal4 DNA­binding domain. The molecular mechanism underlying the inhibition of Notch signaling by a hit compound was further validated using biochemical and cell biological approaches. Using the screening system, a potential candidate, Notch signaling inhibitor­1 (NSI­1), was isolated which showed 50% inhibition at 6.1 µM in an exogenous Notch signaling system. In addition, NSI­1 suppressed the nuclear translocation of NICD and endogenous gene expression of hairy and enhancer of split­1, indicating that NSI­1 specifically targets Notch. Notably, NSI­1 suppressed the cell viability of MCF7 cells and another human breast adenocarcinoma cell line, MDA­MB­231 exhibiting constitutive and high Notch signaling activity, whereas no significant effect was observed in the SH­SY5Y cells bearing a lower Notch signaling activity. NSI­1 significantly suppressed the viability of SH­SY5Y cells expressing exogenous human Notch1. These results indicate that NSI­1 is a novel Notch signaling inhibitor and suggest its potential as a useful drug for the treatment of diseases induced by constitutively active Notch signaling.


Subject(s)
Protein Interaction Domains and Motifs , Receptors, Notch/metabolism , Signal Transduction/drug effects , Cell Line, Tumor , Cell Nucleus , Cell Survival/drug effects , Drug Discovery/methods , Humans , Protein Binding , Protein Transport , Receptors, Notch/chemistry , Receptors, Notch/genetics
17.
EMBO Rep ; 20(7): e46436, 2019 07.
Article in English | MEDLINE | ID: mdl-31267714

ABSTRACT

All vertebrates share a segmented body axis. Segments form from the rostral end of the presomitic mesoderm (PSM) with a periodicity that is regulated by the segmentation clock. The segmentation clock is a molecular oscillator that exhibits dynamic clock gene expression across the PSM with a periodicity that matches somite formation. Notch signalling is crucial to this process. Altering Notch intracellular domain (NICD) stability affects both the clock period and somite size. However, the mechanism by which NICD stability is regulated in this context is unclear. We identified a highly conserved site crucial for NICD recognition by the SCF E3 ligase, which targets NICD for degradation. We demonstrate both CDK1 and CDK2 can phosphorylate NICD in the domain where this crucial residue lies and that NICD levels vary in a cell cycle-dependent manner. Inhibiting CDK1 or CDK2 activity increases NICD levels both in vitro and in vivo, leading to a delay of clock gene oscillations and an increase in somite size.


Subject(s)
Biological Clocks , CDC2 Protein Kinase/metabolism , Cyclin-Dependent Kinase 2/metabolism , Proteolysis , Receptors, Notch/metabolism , Animals , Cell Cycle , Cells, Cultured , Conserved Sequence , Embryonic Stem Cells/metabolism , HEK293 Cells , Humans , Mice , Phosphorylation , Protein Domains , Protein Stability , Receptors, Notch/chemistry
18.
J Cell Physiol ; 234(10): 18448-18458, 2019 08.
Article in English | MEDLINE | ID: mdl-30908631

ABSTRACT

Hepatocellular carcinoma in China accounts for half of the world's incidence. Both epithelial-mesenchymal transition (EMT) and cancer stem cells (CSCs) are thought to be involved in tumor malignant progression. However, the relationship between EMT and CSCs is still unclear. Bioinformatics analysis was performed to evaluate the relationship between EMT and CSCs. The EMT and CSC regulatory mechanism was investigated through Transwell, wound-healing, sphere formation, colony-forming, and western blotting assays. Immunofluorescence and immunoprecipitation were used to study the interaction of hypoxia inducible factor 1α (HIF-1α) /Notch1. Immunohistochemical study was applied to investigate the expression pattern in the process of hepatocellular carcinogenesis and development. In our present study, bioinformatics results indicate that the expression of EMT-related molecules is correlated with CSCs. In vitro studies indicated that EMT activation could induce CSC characteristics. Notch1 was confirmed to mediate the process of EMT-induced CSCs through the interaction with HIF-1α directly. Our findings indicate that EMT could induce CSC-like characteristics, which is mediated by HIF-1α-upregulated Notch intracellular domain expression.


Subject(s)
Carcinoma, Hepatocellular/pathology , Epithelial-Mesenchymal Transition , Liver Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/genetics , Male , Middle Aged , Neoplastic Stem Cells/metabolism , Protein Binding , Protein Domains , Rats, Sprague-Dawley , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Signal Transduction
19.
PLoS Biol ; 17(2): e3000133, 2019 02.
Article in English | MEDLINE | ID: mdl-30716082

ABSTRACT

How multiple receptor tyrosine kinases coordinate cell fate determination is yet to be elucidated. We show here that the receptor for platelet-derived growth factor (PDGF) signaling recruits the p85 subunit of Phosphoinositide 3-kinase (PI3K) to regulate mammalian lens development. Activation of PI3K signaling not only prevents B-cell lymphoma 2 (BCL2)-Associated X (Bax)- and BCL2 Antagonist/Killer (Bak)-mediated apoptosis but also promotes Notch signaling to prevent premature cell differentiation. Reducing PI3K activity destabilizes the Notch intracellular domain, while the constitutive activation of Notch reverses the PI3K deficiency phenotype. In contrast, fibroblast growth factor receptors (FGFRs) recruit Fibroblast Growth Factor Receptor Substrate 2 (Frs2) and Rous sarcoma oncogene (Src) Homology Phosphatase 2 (Shp2) to activate Mitogen-Activated Protein Kinase (MAPK) signaling, which induces the Notch ligand Jagged 1 (Jag1) and promotes cell differentiation. Inactivation of Shp2 restored the proper timing of differentiation in the p85 mutant lens, demonstrating the antagonistic interaction between FGF-induced MAPK and PDGF-induced PI3K signaling. By selective activation of PI3K and MAPK, PDGF and FGF cooperate with and oppose each other to balance progenitor cell maintenance and differentiation.


Subject(s)
Cell Differentiation , Fibroblast Growth Factors/metabolism , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Platelet-Derived Growth Factor/metabolism , Signal Transduction , Animals , Cell Survival , Epithelial Cells/cytology , Epithelial Cells/metabolism , Lens, Crystalline/embryology , Ligands , MAP Kinase Signaling System , Mice , Mutation/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Domains , Protein Stability , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptors, Notch/chemistry , Receptors, Notch/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/genetics
20.
Cell Rep ; 26(4): 845-854.e6, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30673607

ABSTRACT

Notch is a conserved signaling pathway that is essential for metazoan development and homeostasis; dysregulated signaling underlies the pathophysiology of numerous human diseases. Receptor-ligand interactions result in gene expression changes, which are regulated by the transcription factor RBPJ. RBPJ forms a complex with the intracellular domain of the Notch receptor and the coactivator Mastermind to activate transcription, but it can also function as a repressor by interacting with corepressor proteins. Here, we determine the structure of RBPJ bound to the corepressor SHARP and DNA, revealing its mode of binding to RBPJ. We tested structure-based mutants in biophysical and biochemical-cellular assays to characterize the role of RBPJ as a repressor, clearly demonstrating that RBPJ mutants deficient for SHARP binding are incapable of repressing transcription of genes responsive to Notch signaling in cells. Altogether, our structure-function studies provide significant insights into the repressor function of RBPJ.


Subject(s)
DNA-Binding Proteins , Immunoglobulin J Recombination Signal Sequence-Binding Protein , Multiprotein Complexes , RNA-Binding Proteins , Signal Transduction , Transcription, Genetic , Animals , Binding Sites , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/chemistry , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Structure, Quaternary , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Notch/chemistry , Receptors, Notch/genetics , Receptors, Notch/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...