scholarly journals Long-range Notch-mediated tissue patterning requires actomyosin contractility

2018 ◽  
Author(s):  
Ginger L Hunter ◽  
Li He ◽  
Norbert Perrimon ◽  
Guillaume Charras ◽  
Edward Giniger ◽  
...  

AbstractDynamic, actin-based protrusions function in cell-cell signaling in a variety of systems. In the dorsal thorax of the developing fly, basal protrusions enable non-neighboring epithelial cells to touch, extending the range over which Notch-mediated lateral inhibition can occur during bristle patterning. Given that actin-based cell protrusions can exert mechanical forces on their environment and Notch receptor activation is mechanically sensitive, how might cytoskeletal contractility contribute to Notch signaling? We identify a pool of basal non-muscle myosin II (myosin II) that regulates protrusion dynamics, promotes Notch signaling, and is required in signal sending and receiving cells for Notch-dependent patterning. We show that interactions between protrusions are extensive and subject to actomyosin contractility. The effects of reducing myosin II activity are more pronounced for protrusion-mediated signaling than for signaling at lateral cell contacts. Together, these results reveal a role for actomyosin contractility in Notch activation, signaling, and patterning in a developmental context.

Biomolecules ◽  
2021 ◽  
Vol 11 (2) ◽  
pp. 309
Author(s):  
Wataru Saiki ◽  
Chenyu Ma ◽  
Tetsuya Okajima ◽  
Hideyuki Takeuchi

The 100th anniversary of Notch discovery in Drosophila has recently passed. The Notch is evolutionarily conserved from Drosophila to humans. The discovery of human-specific Notch genes has led to a better understanding of Notch signaling in development and diseases and will continue to stimulate further research in the future. Notch receptors are responsible for cell-to-cell signaling. They are activated by cell-surface ligands located on adjacent cells. Notch activation plays an important role in determining the fate of cells, and dysregulation of Notch signaling results in numerous human diseases. Notch receptors are primarily activated by ligand binding. Many studies in various fields including genetics, developmental biology, biochemistry, and structural biology conducted over the past two decades have revealed that the activation of the Notch receptor is regulated by unique glycan modifications. Such modifications include O-fucose, O-glucose, and O-N-acetylglucosamine (GlcNAc) on epidermal growth factor-like (EGF) repeats located consecutively in the extracellular domain of Notch receptors. Being fine-tuned by glycans is an important property of Notch receptors. In this review article, we summarize the latest findings on the regulation of Notch activation by glycosylation and discuss future challenges.


2013 ◽  
Vol 210 (2) ◽  
pp. 321-337 ◽  
Author(s):  
Sankaranarayanan Kannan ◽  
Robert M. Sutphin ◽  
Mandy G. Hall ◽  
Leonard S. Golfman ◽  
Wendy Fang ◽  
...  

Although aberrant Notch activation contributes to leukemogenesis in T cells, its role in acute myelogenous leukemia (AML) remains unclear. Here, we report that human AML samples have robust expression of Notch receptors; however, Notch receptor activation and expression of downstream Notch targets are remarkably low, suggesting that Notch is present but not constitutively activated in human AML. The functional role of these Notch receptors in AML is not known. Induced activation through any of the Notch receptors (Notch1–4), or through the Notch target Hairy/Enhancer of Split 1 (HES1), consistently leads to AML growth arrest and caspase-dependent apoptosis, which are associated with B cell lymphoma 2 (BCL2) loss and enhanced p53/p21 expression. These effects were dependent on the HES1 repressor domain and were rescued through reexpression of BCL2. Importantly, activated Notch1, Notch2, and HES1 all led to inhibited AML growth in vivo, and Notch inhibition via dnMAML enhanced proliferation in vivo, thus revealing the physiological inhibition of AML growth in vivo in response to Notch signaling. As a novel therapeutic approach, we used a Notch agonist peptide that led to significant apoptosis in AML patient samples. In conclusion, we report consistent Notch-mediated growth arrest and apoptosis in human AML, and propose the development of Notch agonists as a potential therapeutic approach in AML.


2021 ◽  
Author(s):  
Miho Matsuda ◽  
Chih-Wen Chu ◽  
Sergei S Sokol

The reduction of the apical domain, or apical constriction, is a process that occurs in a single cell or is coordinated in a group of cells in the epithelium. Coordinated apical constriction is particularly important when the epithelium is undergoing dynamic morphogenetic events such as furrow or tube formation. However, the underlying mechanisms remain incompletely understood. Here we show that Lim only protein 7 (Lmo7) is a novel activator of apical constriction in the Xenopus superficial ectoderm, which coordinates actomyosin contractility in a group of cells during epithelial morphogenesis. Like other apical constriction regulators, Lmo7 requires the activation of the Rho-Rock-Myosin II pathway to induce apical constriction. However, instead of increasing the phosphorylation of myosin light chain (MLC), Lmo7 binds muscle myosin II heavy chain A (NMIIA) and increases its association with actomyosin bundles at adherens junctions (AJs). Lmo7 overexpression modulates the subcellular distribution of Wtip, a tension marker at AJs, suggesting that Lmo7 generates mechanical forces at AJs. We propose that Lmo7 increases actomyosin contractility at AJs by promoting the formation of actomyosin bundles.


Blood ◽  
2010 ◽  
Vol 115 (14) ◽  
pp. 2777-2783 ◽  
Author(s):  
Julien Y. Bertrand ◽  
Jennifer L. Cisson ◽  
David L. Stachura ◽  
David Traver

Abstract Recent studies have revealed that definitive hematopoiesis in vertebrates initiates through the formation of a non–self-renewing progenitor with limited multilineage differentiation potential termed the erythromyeloid progenitor (EMP). EMPs are specified before hematopoietic stem cells (HSCs), which self-renew and are capable of forming all mature adult blood lineages including lymphoid cells. Despite their differences, EMPs and HSCs share many phenotypic traits, making precise study of their respective functions difficult. Here, we examine whether embryonic specification of EMPs requires Notch signaling as has been shown for HSCs. In mindbomb mutants, which lack functional Notch ligands, we show that EMPs are specified normally: we detect no significant differences in cell number, gene expression, or differentiation capacity between EMPs purified from wild-type (WT) or mindbomb mutant embryos. Similarly N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT), a chemical inhibitor of Notch receptor activation, has no effect on EMP specification. These studies establish that HSCs are the only hematopoietic precursor that requires Notch signaling and help to clarify the signaling events underlying the specification of the 2 distinct waves of definitive hematopoiesis.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Bettina Kunze ◽  
Moritz Middelhoff ◽  
H. Carlo Maurer ◽  
Tatiana Agibalova ◽  
Akanksha Anand ◽  
...  

AbstractBarrett’s esophagus (BE) is a precursor to esophageal adenocarcinoma (EAC), but its cellular origin and mechanism of neoplastic progression remain unresolved. Notch signaling, which plays a key role in regulating intestinal stem cell maintenance, has been implicated in a number of cancers. The kinase Dclk1 labels epithelial post-mitotic tuft cells at the squamo-columnar junction (SCJ), and has also been proposed to contribute to epithelial tumor growth. Here, we find that genetic activation of intracellular Notch signaling in epithelial Dclk1-positive tuft cells resulted in the accelerated development of metaplasia and dysplasia in a mouse model of BE (pL2.Dclk1.N2IC mice). In contrast, genetic ablation of Notch receptor 2 in Dclk1-positive cells delayed BE progression (pL2.Dclk1.N2fl mice), and led to increased secretory cell differentiation. The accelerated BE progression in pL2.Dclk1.N2IC mice correlated with changes to the transcriptomic landscape, most notably for the activation of oncogenic, proliferative pathways in BE tissues, in contrast to upregulated Wnt signalling in pL2.Dclk1.N2fl mice. Collectively, our data show that Notch activation in Dclk1-positive tuft cells in the gastric cardia can contribute to BE development.


Biomolecules ◽  
2021 ◽  
Vol 11 (9) ◽  
pp. 1369 ◽  
Author(s):  
Judith Hounjet ◽  
Marc Vooijs

Aberrant Notch signaling has been found in a broad range of human malignancies. Consequently, small molecule inhibitors and antibodies targeting Notch signaling in human cancers have been developed and tested; however, these have failed due to limited anti-tumor efficacy because of dose-limiting toxicities in normal tissues. Therefore, there is an unmet need to discover novel regulators of malignant Notch signaling, which do not affect Notch signaling in healthy tissues. This review provides a comprehensive overview of the current knowledge on the role of intracellular trafficking in ligand-independent Notch receptor activation, the possible mechanisms involved, and possible therapeutic opportunities for inhibitors of intracellular trafficking in Notch targeting.


Blood ◽  
2009 ◽  
Vol 113 (13) ◽  
pp. 3092-3101 ◽  
Author(s):  
Amy L. Radke ◽  
Lauren E. Reynolds ◽  
Rossana C. N. Melo ◽  
Ann M. Dvorak ◽  
Peter F. Weller ◽  
...  

Abstract Eosinophil chemotaxis and survival within tissues are key components in the development of tissue eosinophilia and subsequent effector responses. In this study, we demonstrate a novel mechanism of eosinophil autoregulation affecting migration and survival mediated through Notch signaling. We show for the first time that human blood eosinophils express Notch receptors and Notch ligands, expressions of which are influenced by the presence of eosinophil-activating granulocyte-macrophage colony-stimulating factor (GM-CSF). Evidence of Notch receptor activation and subsequent transcription of the Notch-responsive gene HES1 were observed in GM-CSF–stimulated eosinophils, confirming functionality of eosinophil-expressed Notch-signaling components. Moreover, by inhibiting Notch signaling with γ-secretase inhibitors or Notch receptor–specific neutralizing antibodies, we demonstrate that autocrine Notch signaling enhances stimulus-mediated actin rearrangement and eosinophil chemokinesis, and impairs eosinophil viability. Taken together, these data suggest autocrine Notch signaling, enhanced in response to tissue- or inflammatory-derived signals, influences eosinophil activity and longevity, which may ultimately contribute to the development of tissue eosinophilia and exacerbation or remediation of eosinophil effector functions.


Biomolecules ◽  
2021 ◽  
Vol 11 (11) ◽  
pp. 1672
Author(s):  
Anja C. Nagel ◽  
Dominik Müller ◽  
Mirjam Zimmermann ◽  
Anette Preiss

The Notch signaling pathway is pivotal to cellular differentiation. Activation of this pathway involves proteolysis of the Notch receptor and the release of the biologically active Notch intracellular domain, acting as a transcriptional co-activator of Notch target genes. While the regulation of Notch signaling dynamics at the level of ligand–receptor interaction, endocytosis, and transcriptional regulation has been well studied, little is known about factors influencing Notch cleavage. We identified EP555 as a suppressor of the Notch antagonist Hairless (H). EP555 drives expression of CG32521 encoding membrane-bound proteins, which we accordingly rename membrane-bound Notch regulator (mnr). Within the signal-receiving cell, upregulation of Mnr stimulates Notch receptor activation, whereas a knockdown reduces it, without apparent influence on ligand–receptor interaction. We provide evidence that Mnr plays a role in γ-secretase-mediated intramembrane cleavage of the Notch receptor. As revealed by a fly-eye-based reporter system, γ-secretase activity is stimulated by the overexpression of Mnr, and is inhibited by its knockdown. We conclude that Mnr proteins support Notch signaling activity by fostering the cleavage of the Notch receptor. With Mnr, we identified a membrane-bound factor directly augmenting Notch intra-membrane processing, thereby acting as a positive regulator of Notch signaling activity.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 463-463
Author(s):  
Michelle I Lin ◽  
Emily Price ◽  
Sonja Boatman ◽  
Eirini Trompouki ◽  
Sruthi Satishchandran ◽  
...  

The angiopoietin-like proteins (angptls) are novel growth factors that are capable of stimulating expansion of mouse and human hematopoietic stem and progenitor cells (HSPCs) ex vivo. Although the receptor for some of the angptl family members is known, their molecular mechanism of action is undefined. Here, we show that overexpression of angptl2 in a stable heatshock inducible transgenic zebrafish line, Tg(hsp70:zangptl2), is sufficient to increase cmyb- and runx1-positive HSPCs in the aorta-gonad-mesonephros (AGM) region, the site of definitive hematopoiesis. Anti-sense morpholino knockdown of angptl1 and 2 resulted in a significant decrease in cmyb- and runx1-positive HSPCs in the AGM, suggesting that angptls are required for definitive hematopoiesis. These double morphants also displayed severe disruption in vascular development and differentiation prior to the defects observed in the AGM, indicating that angptl regulation of HSPC development occurs through an early specification of the hemogenic endothelium. The loss of function phenotypes in the developing aorta is reminiscent of mutant fish with defective notch signaling (mindbomb) and a tight genetic interaction occurs between angptl and notch signaling. Knocking down angptl1 and 2 decreased notch signaling in a transgenic notch reporter line while overexpression of a constitutively active intracellular notch rescues the angptl double morphant phenotype. These data imply that angptls function upstream of notch signaling. Interestingly, the absence of HSPCs and notch signaling in the mindbomb mutants are rescued by overexpression of angptl2 in Tg(hsp70:zangptl2), suggesting that angptls can regulate notch signaling. To examine the molecular mechanism of Angptl-mediated Notch activation, we stimulated cultured human CD34+ cells or endothelial cells with purified Angptl2 and observed a rapid increase in Notch receptor cleavage indicating that Angptl2 can induce Notch activation. Furthermore, we found through endogenous co-immunoprecipitation experiments that the Angptl receptor, LILRB2, interacted with Notch receptor. This strongly points to a direct regulation of Notch activation/signaling by Angptls through physical interactions between Notch and Angptl receptors. Previously, we found that angptl-mediated akt activation is important for HSPC formation in the AGM. To examine the role of Akt during Notch activation, we immunoprecipitated all Akt-phosphorylated substrates in Angptl-stimulated cells and found the presence of ADAM17/TACE, one of the cleavage enzymes that is crucial to Notch receptor activation. Together these results suggest a model in which Angptl-binding of the LILRB2 receptor enables recruitment of downstream molecules such as Akt proximal to Notch, allowing for subsequent cleavage and activation of Notch receptor. Finally, to examine downstream signaling of Angptl-mediated Notch activation, we performed chromatin immunoprecipitation for Notch followed by sequencing in Angptl2-stimulated CD34+ cells and found enrichment for Myc binding elements. Independent microarrays also revealed a strong Myc signature through gene set enrichment analysis. Thus, to confirm results from our bioinformatics analyses, we overexpressed zebrafish myc in angptl1 and 2 double morphants or mindbomb mutants, and found a significant rescue in HSPCs formation in the AGM. Collectively with these results, we propose that angptl can regulate notch signaling through receptor interaction, leading to activation of myc target genes during definitive hematopoiesis. Our data provide new insights to the previously uncharacterized Angptl signaling during HSPC development and present a novel mechanism of action for Notch activation. Disclosures: Aster: Cell Signaling Technology: Consultancy; Merck, Inc.: Research Funding; Pfizer, Inc.: Research Funding; Genentech, Inc.: Honoraria. White:N-of-One: Consultancy, Equity Ownership. Zon:FATE Therapeutics, Inc: Consultancy, Equity Ownership, Founder Other, Membership on an entity’s Board of Directors or advisory committees, Patents & Royalties; Stemgent, Inc: Consultancy, Membership on an entity’s Board of Directors or advisory committees, Stocks, Stocks Other; Scholar Rock: Consultancy, Equity Ownership, Founder, Founder Other, Membership on an entity’s Board of Directors or advisory committees, Patents & Royalties.


2006 ◽  
Vol 173 (3) ◽  
pp. 443-452 ◽  
Author(s):  
Elliott J. Hagedorn ◽  
Jennifer L. Bayraktar ◽  
Vasundhara R. Kandachar ◽  
Ting Bai ◽  
Dane M. Englert ◽  
...  

We have isolated mutations in the Drosophila melanogaster homologue of auxilin, a J-domain–containing protein known to cooperate with Hsc70 in the disassembly of clathrin coats from clathrin-coated vesicles in vitro. Consistent with this biochemical role, animals with reduced auxilin function exhibit genetic interactions with Hsc70 and clathrin. Interestingly, the auxilin mutations interact specifically with Notch and disrupt several Notch-mediated processes. Genetic evidence places auxilin function in the signal-sending cells, upstream of Notch receptor activation, suggesting that the relevant cargo for this auxilin-mediated endocytosis is the Notch ligand Delta. Indeed, the localization of Delta protein is disrupted in auxilin mutant tissues. Thus, our data suggest that auxilin is an integral component of the Notch signaling pathway, participating in the ubiquitin-dependent endocytosis of Delta. Furthermore, the fact that auxilin is required for Notch signaling suggests that ligand endocytosis in the signal-sending cells needs to proceed past coat disassembly to activate Notch.


Sign in / Sign up

Export Citation Format

Share Document