Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Mar 16.
Published in final edited form as: Curr Alzheimer Res. 2012 Feb;9(2):227–240. doi: 10.2174/156720512799361600

An Overview of Notch Signaling in Adult Tissue Renewal and Maintenance

Chihiro Sato 1, Guojun Zhao 1, Ma Xenia G Ilagan 1,*
PMCID: PMC4361071  NIHMSID: NIHMS660579  PMID: 21605032

Abstract

The Notch pathway is a critical mediator of short-range cell-cell communication that is reiteratively used to regulate a diverse array of cellular processes during embryonic development and the renewal and maintenance of adult tissues. Most Notch-dependent processes utilize a core signaling mechanism that is dependent on regulated intramembrane proteolysis: Upon ligand binding, Notch receptors undergo ectodomain shedding by ADAM metalloproteases, followed by γ-secretase-mediated intramembrane proteolysis. This releases the Notch intracellular domain, which translocates to the nucleus to activate transcription. In this review, we highlight the roles of Notch signaling particularly in self-renewing tissues in adults and several human diseases and raise some key considerations when targeting ADAMs and γ-secretase as disease-modifying strategies for Alzheimer's Disease.

Keywords: ADAM, CADASIL, γ-secretase, multiple sclerosis, Notch, T-ALL, stem cell, tissue renewal

Overview of the Notch signaling pathway

The Notch pathway is one of several conserved signaling pathways that regulate cell proliferation, cell fate decisions and induction of differentiation during embryonic and postnatal development [1-4]. The Notch receptors (Notch1-4 in mammals) are large Type I transmembrane proteins (Fig 1a). The extracellular domain is composed of 29-36 EGF repeats, three conserved cysteine-rich Lin12-Notch repeats (LNR) and the heterodimerization domain (HD). Together, the LNR and heterodimerization domains constitute the Negative regulatory region (NRR), which functions to keep the receptor “off” in the absence of ligand [5-9]. The single transmembrane domain (TMD) is then followed by a large intracellular region, which contains the RAM (RBPjκ association module) domain, nuclear localization signals (NLS), seven ankyrin repeats (ANK) and the PEST (Proline/GLutamic acid/Serine/Threonine-rich) domain.

Figure 1.

Figure 1

Overview of the Notch signaling pathway.

(a) Schematic depiction of mammalian Notch receptors and DSL ligands. Notch receptors are large single-pass Type I membrane proteins. The extracellular domain has 29-36 epidermal growth factor (EGF) repeats, followed by the conserved Lin12-Notch repeats (LNR: light green) and a heterodimerization domain (HD). The LNR and HD domains constitute the Negative regulatory region (NRR). The single transmembrane domain (TMD: pink) is followed by the RBPjκ association module (RAM) domain (purple), nuclear localization sequences (NLSs: blue), 7 ankyrin repeats (ANK) (light blue), and a transactivation domain containing a C-terminal proline/glutamic acid/serine threonine-rich motifs (PEST: gray). The major Notch ligands are also Type I membrane proteins that have an N-terminal Delta/Serrate/LAG-2 (DSL: light pink) domain and multiple EGF motifs (light blue). The ligands can be further classified based on the presence or absence of the Cysteine-rich (CR; purple) and of the Delta and OSM-11-like protein (DOS; lavender) domains.

(b) Canonical Notch signaling pathway. Notch receptors are expressed at the plasma membrane as intramolecular heterodimers, held together by non-covalent interactions within the heterodimerization domain (HD). The receptors are activated by binding to ligands presented by a neighboring signal-sending cell. Ligand endocytosis is thought to generate the force needed to unfold the NRR (and perhaps dissociate the HD) to expose S2 to ADAM cleavage. Ectodomain shedding generates the membrane-anchored intermediate NEXT (Notch extracellular truncation), which is subsequently cleaved by γ-secretase progressively within the TMD to release the Notch intracellular domain (NICD) and the Nβ peptides. γ-secretase cleavage of Notch can occur at the cell surface or in an endosomal compartment. Once released, NICD translocates to the nucleus, associates with DNA-binding protein CSL and the transcriptional coactivator Mastermind to activate transcription. Some Notch ligands also go through sequential cleavages by ADAM proteases and γ-secretase. ADAM-mediated shedding of ligand ectodomains has been shown to modulate the strength and timing of Notch pathway activity. The role of the γ-secretase cleavage of ligands and released ligand intracellular domain fragments is still unclear.

(c) TMD region of mouse Notch1, showing cleavage sites and products. Notch is cleaved by an ADAM protease at the S2 site. After ectodomain shedding, γ-secretase cleaves the Notch TMD, starting at the S3 site. Multiple scissile bonds can be cleaved to release different NICD molecules with varying N-terminal residues. Only NICD-V (blue) is resistant to N-end rule degradation and is likely the major biologically functional species. TMD proteolysis then proceeds toward S4 until the Nβ peptides can be released.

Most receptors undergo processing at cleavage site1 (S1) by furin-like proteases in the Golgi and are therefore targeted to the cell surface as an intramolecular heterodimer held together by non-covalent interactions between the N- and C-terminal HD regions (Fig 1b). Recent studies using structure-guided deletions of the S1 sites in human Notch1 and Notch2 demonstrated that S1 cleavage is not absolutely required for receptor activation but it can have differential effects on cell surface trafficking [10], suggesting that this could be a mechanism for regulating availability at the cell surface (and thereby, activation potential) of different paralogs.

The Notch receptor is activated by binding to a ligand presented by a neighboring cell (Fig 1b). The major Notch ligands are also Type I transmembrane proteins that are characterized by an N-terminal DSL (Delta/Serrate/Jagged/Lag-2) domain and can be further classified based on the presence or absence of a cysteine-rich domain (Serrate/Jagged ligands have this domain, Delta ligands do not) (Fig 1a) [9, 11]. The mechanistic details of how ligand binding promotes Notch receptor proteolysis and activation are emerging (Fig 1b) [7, 9, 12]. Ligand endocytosis is thought to be required for ligand maturation [13-15] and also, after binding to the receptor, to generate sufficient force to produce a conformational change in the NRR [5, 8, 16]. The unfolding of the NRR (perhaps following heterodimer dissociation at S1 [17]) will expose site 2 (S2) to proteolysis by ADAM10/Kuz, the major metalloprotease involved in S2 cleavage of ligand-activated Notch receptors [18, 19]. This is consistent with the ADAM10/Kuz/sup-17 loss-of-function phenotypes (but not those of ADAM17/TACE) phenocopying the defects associated with Notch deficiency [20-22]. However, other metalloproteases may also play a role in tissues where ADAM10 is not expressed and in disease states. Elevated metalloprotease levels can lead to ligand-independent activation of Notch receptors [23, 24]. Interestingly, Notch receptors that are activated in a ligand-independent manner (specifically, by calcium chelation [25], or by T-ALL-associated and other activating mutations that destabilize the NRR [26, 27]) are no longer mainly dependent on ADAM10 for ectodomain shedding. ADAM17/TACE (and perhaps other metalloproteases) appears to play a more prominent role under these conditions [18, 19]. The molecular mechanisms underlying this selectivity are still unclear. This could reflect differences in structures between the various ADAMs and/or differences in the conformational changes of the receptor under different modes of receptor activation. Differential spatial distribution of the various ADAMs within the cell (relative to γ-secretase, for instance) may also be critical.

S2 cleavage is a major regulatory step in the Notch activation process. Notch ectodomain shedding creates the membrane-anchored intermediate fragment NEXT (Notch extacellular truncation), which is a direct substrate for the γ-secretase enzyme (Fig 1b). γ-Secretase is a multi-component intramembrane-cleaving protease composed of the catalytic component Presenilin (PS), along with Nicastrin, Pen2 and Aph1 [28, 29]. The γ-secretase complex initially cleaves the Notch TMD at the cytoplasmic end (site S3), which releases the Notch intracellular domain (NICD). Progressive cleavages towards the middle of the TMD eventually release the Nβ peptides (from site S4) (Fig 1c) [30]. The precise cellular location where γ-secretase cleaves Notch remains a subject of intense investigation [3, 9, 15]. Early genetic studies demonstrated the requirement of endocytosis in both the signal-sending and signal-receiving cells [16, 31]. As mentioned earlier, endocytosis of ligand during the activation process could be providing the force to unfold the NRR for exposure of S2 to ADAM. In the signal-receiving cell, endocytosis could also be contributing to this process [9, 32]. Alternatively or in addition, endocytosis might be required for cleavage to occur (at least in some contexts) [33]. γ-Secretase is active in many cellular membranes and studies have shown that γ-secretase cleavage of Notch can occur at the cell surface or in an endosomal compartment (Fig 1b). Moreover, the cellular location where cleavage occurs can influence the precise position of S3 cleavage and as a consequence, modulate the stability of released NICD molecules [34]. This too can potentially be a point of pathway regulation. After NICD is released by γ-secretase, it travels to the nucleus where it associates with the DNA-binding protein CSL (CBF-1/RBPjκ, Su(H), Lag-1). Together, they recruit the coactivator Mastermind, and further assemble a transcriptional complex that activates downstream targets (Fig 1b) [2, 7, 9, 35, 36].

Interestingly, Notch ligands also undergo sequential proteolytic processing by ADAMs and γ-secretase (Fig 1b) [11, 37]. Ligand proteolysis can be constitutive and likely depends on the availability of ADAMs. Indeed, activating ADAMs with p-aminophenylmercuric acetate increases ligand cleavage [38]. Notch receptor binding can also enhance ligand cleavage [38, 39]. ADAM-mediated ectodomain shedding of ligands is thought to be important for their downregulation, which helps promote and maintain unidirectional signaling [38, 40-42] or alleviate cis-inhibition [41, 43]. Ligand ectodomain shedding by ADAM appears to be dispensable during T-cell development [44] but is critical during cortical neurogenesis [45] and for maintaining the balance between self-renewal and differentiation in muscle satellite cells [46]. In contrast, the functional significance of the γ-secretase cleavage of ligands is less clear. γ-Secretase cleavage might just be facilitating disposal of the membrane-tethered carboxy-terminal fragments that are left after ADAM-mediated shedding [47, 48]. Alternatively, ligand proteolysis could be releasing functional ICD fragments (analogous to NICD) that could regulate transcription and be involved in ligand back signaling [39, 49]. Because most of the studies regarding the functions of released ligand ICD fragments have relied on overexpressed or engineered protein fragments, additional studies will have to address whether such a bidirectional mode of signaling from endogenously expressed and cleaved ligands plays a role in specific contexts in vivo. Notably, γ-secretase-deficient ligand-expressing cells appear to signal well for most Notch-mediated decisions, and display no obvious phenotypes [50, 51].

The core or canonical pathway is conserved for all Notch receptors and is utilized in most Notch-dependent processes. This direct membrane-to-nucleus signaling mechanism occurs without any signal amplification by secondary messengers, but is regulated by post-translational modifications (e.g., glycosylation, phosphorylation, ubiquitination) and endocytic trafficking of both ligands and receptors [2, 3, 11, 14, 15, 52-57]. In a context-dependent manner, these regulatory mechanisms modulate the strength and timing of Notch signals, and ultimately their biological consequences.

The essential function of Notch signaling in development is evident in the early embryonic lethality associated with loss of Notch signaling. Global knockouts of Notch1 or 2 in mice are embryonic lethal mainly due to the early defects in neurons, somites, and vascular integrity both in the embryo and placenta [58-61]. In addition, mice homozygous for the Notch1 V1744G allele, which produce highly unstable NICD molecules, phenocopy the Notch1 null mice [34, 62]. Ablation of RBPjκ abolishes all canonical Notch signaling and RBPjκ global knockout mice exhibit similar or more severe phenotypes than those of Notch1 null embryos [63]. Knockouts of the other key components and modifiers of the Notch signaling pathway also support the view that Notch signaling is dispensable for gastrulation but essential for post-implantation mammalian development, starting at E9 [64]. It is important to note in this context that ablation of γ-secretase components, especially PS1, is almost identical to Notch-deficient mice [65, 66]. PS2 is dispensable for embryogenesis, but the double knockout of PS1 and PS2 is more severe and nearly identical to Notch signaling knockout mice [67]. Nicastrin- and Aph1a-null mice also exhibit embryonic lethal phenotypes, although Aph1b and c are dispensable for embryogenesis [68-72]. These studies strongly suggest that γ-secretase inhibition can phenocopy the loss of Notch signaling, and confirms the central role of γ-secretase as a mediator of the Notch signaling mechanism.

A growing number of studies have begun to investigate the roles of Notch signaling in adult mammals, thanks to the recent advances in inducible Cre-loxP targeting technology, which makes it possible to study both temporal and spatial regulation and functions of Notch signaling in vivo. This review will primarily focus on the biological role of Notch signaling in adult mammals in an attempt to delineate the possible complications that could be caused by γ-secretase inhibition and/or modulation in Alzheimer's disease (AD) therapy. In addition, we wish to identify other possible clinical applications wherein Notch modulation by γ-secretase inhibitors (GSIs) might be useful.

Notch Signaling in Adult Tissue Renewal and Maintenance

It has been demonstrated that Notch signaling is critical in tissue renewal and maintenance in various organs, including but not limited to, the intestine, skin, blood, liver, kidney, nervous system, bone and muscle. Thus, one can easily imagine that acute or chronic disruption of the pathway can lead to various complications in multiple organs. However, important questions such as how much reduction of Notch signaling can be tolerated in each tissue remain largely unanswered. In addition, Notch signaling functions in a highly context-dependent manner; biological consequences of pathway activation can vary depending on multiple factors, such as cell type, timing, and mode of signaling. Therefore, understanding the role of Notch signaling in each organ is critical for identifying a therapeutic window for GSIs. In the following sections, we highlight some of the key roles of Notch signaling in actively self-renewing tissues, such as the intestine, hematopoietic system, and skin. In addition, we discuss the potential roles of Notch signaling in cell renewal systems within the brain and muscle, which go through slower turnover.

Notch signaling in adult intestine

The epithelium of the intestine is one of the most rapidly and dynamically renewed tissues in the adult. It renews every four to five days, and failure to maintain this homeostasis can lead to death. Mild disruptions can lead to malnutrition, infection, and cancer. Cell renewal of the epithelium relies heavily on its proliferating LGR5+ intestinal stem cells (ISCs), which reside at the bottom of the crypt, and quiescent +4 position, Bmi+ ISCs [73]. LGR5+ cells first divide symmetrically to stochastically generate transit amplifying (TA) cells [74, 75], which upon leaving the TA compartment at the crypt-villus junction, subsequently give rise to all four different terminally differentiated cells: absorptive enterocyte, secretory goblet cells, enteroendocrine, and Paneth cells (Fig 2a) [76-79]. Notch signaling has been shown to regulate cell renewal and binary fate decisions in the adult intestine in concert with Wnt signaling, which functions as the master switch that promotes cell proliferation and suppresses differentiation (Fig 2a) [77-83]. Ablation of Notch signaling in the intestine using conditional Notch1/Notch2 double knockout, or conditional RBPjκ knockout mice, or the GSI dibenzazepine, was shown to specifically increase the number of secretory goblet cells at the expense of absorptive enterocyte cells [84, 85]. This suggests that Notch signaling regulates binary cell fate decision between these two cell lineages. In a reciprocal experiment, forced expression of NICD in the intestine led to reduction of secretory cells and increased cell proliferation [86]. These results suggest that there are two roles of Notch signaling in the adult intestine: it promotes proliferation either in the ISC or TA cell compartment, and it regulates binary fate decisions between absorptive and secretory cells. The first role was shown to be Wnt signaling-dependent while the second is independent of Wnt signaling [87]. In zebrafish, lateral inhibition through Delta-mediated Notch signaling has been implicated to play a role in these binary cell fate decisions [88]. Notch1 and Notch2 have been shown to function redundantly in the gut [84, 89].

Figure 2.

Figure 2

Notch signaling is utilized in multiple organs in the adult for cell renewal and tissue maintenance.

(a) In the adult intestine, there are two types of adult intestinal stem cells (ISCs): Bmi+ quiescent and Lgr5+ proliferating ISCs (both in pink). They produce transit amplifying (TA) cells (light orange), and differentiate into four types of cells: absorptive enterocyte (light blue), secretory goblet (green), enteroendocrine (blue), and Paneth (purple) cells. Notch signaling promotes (blue arrow) proliferation of the adult ISCs and determines binary cell fate decisions between absorptive and secretory cells.

(b) During adult hematopoiesis, Notch signaling promotes T cell lineage differentiation and regulates multiple other differentiation steps. It is still controversial whether Notch signaling regulates proliferation of hematopoietic stem cells (HSCs: pink) in the adult. Leukemia/lymphoma related factor (LRF) is a negative regulator of Notch signaling in the bone marrow.

(c) In the adult skin, Notch signaling promotes spinous cell fate. Ablation of Notch signaling leads to tumorigenesis through non-cell autonomous effects. It also leads to secretion of thymic stromal lymphopoietin (TSLP), which can eventually lead to atopic dermatitis and asthma in the adult.

(d) Adult neurogenesis takes place in two regions of the brain, the subgranular zone (SGZ) of the hippocampus and the subventicular zone (SVZ). In the SVZ, Notch signaling promotes cell proliferation in the adult neural stem cells (NSCs, pink, marked by glial fibrillary acidic protein (GFAP), glutamate-aspartate transporter (GLAST), or Nestin) and inhibits differentiation toward transit amplifying (TA) cells (light blue). Notch signaling maintains quiescence of ependymal cells (ciliated cells), which produce astrocyte (pink) and neuroblast (blue) upon stroke/injury. In the SGZ, Notch signaling promotes proliferation of adult NSCs (pink) and inhibits entry from NSCs to TA cells and/or cell cycle exit. Notch signaling could also promote survival and maturation of neurons and modulate neuritogenesis in mature neurons.

(e) In adult muscle, Notch signaling is activated upon injury in satellite cells (light blue), which are the adult muscle stem cells, and promotes proliferation to produce myogenic progenitors (pink). Asymmetric localization of Notch inhibitor Numb during cell divisions results in two daughter cells: myoblast (blue) and a myogenic progenitor.

Notch activation in the intestine was also shown to promote tumorigenesis in a sensitized background (Apcmin/+) that has a propensity for developing intestinal adenomas [87]. Treatment with GSIs promoted goblet cell differentiation and reduced proliferation in adenomas [85], suggesting that GSIs may also be useful in the treatment of neoplastic diseases in the gut [90]. However, severe gut toxicity has already been demonstrated in multiple independent rodent toxicological studies with GSIs [85, 91-93], suggesting that dosage or regional control of γ-secretase inhibition is critical. Recent in vivo studies using a combination of the GSI compound E, and the glucocorticoid dexamethasone, have successfully improved the anti-leukemic effects in T-ALL while reducing intestinal toxicity, showing that dosage control is feasible [94].

Notch signaling in adult hematopoiesis

The hematopoietic system undergoes rapid and robust cell renewal throughout life, and its dysregulation can lead to various hematologic diseases such as anemia, leukemia, and immune disorders. During embryonic development, hematopoietic stem cells (HSCs) primitively arise from the yolk sac and aorta-gonad mesonephros, which is followed by its generation in the fetal liver and bone marrow. In adult animals, HSCs primarily reside in the bone marrow, and they can give rise to all hematopoietic cell lineages, including myeloid cells (monocytes, macrophages, neutrophils, basophils, eosinophils, erythrocytes, megakaryocytes, dendritic cells) and lymphoid cells (T cells, B cells, natural killer (NK) cells) [95, 96].

Notch signaling has been implicated in multiple cell fate decisions during hematopoiesis (Fig 2b) [80, 97-99]. Examples include cell differentiation decisions toward marginal zone B/follicular B cell [100, 101], CD8+ cytotoxic T cell [102, 103], Th1/Th2 cell [104], megakaryocyte [105], mast cell [106], NK cell [107, 108], and dendritic cell [109, 110] fates, but the best-studied role of Notch signaling is in T cell lineage commitment owing to the discovery of its causative role in human T cell acute lymphoblastic leukemia (T-ALL), which will be further discussed later. A number of loss- and gain-of-function studies of Notch1 in bone marrow progenitors have demonstrated that Notch signaling instructs T cell fate at the expense of B cell fate [111-114]. Importantly, early versions of GSIs inhibited T cell development in mouse fetal thymus organ cultures [115, 116]. In follow up studies with human-mouse fetal thymus organ cultures, the GSI DAPT was shown to dose-dependently affect different hematopoietic cell lineage development: T cells required a higher dose of Notch signaling, NK cells were generated with a low dosage of Notch signaling whereas B-cells were generated in the absence of Notch signaling [117]. T cell lineage commitment was shown to be mediated through Dll4 expressed in thymic epithelial cells, implicating a thymic niche for T cell development [118, 119]. Proto-oncogene leukemia/lymphoma related factor (LRF) was identified as a negative regulator of Notch signaling in bone marrow progenitors, possibly providing a mechanism for suppressing the level of Notch signaling in order to prevent ectopic differentiation into T cell lineages [120]. It has been further shown that Notch signaling inhibits additional cell fate decisions including myeloid cells and dendritic cells to ensure T cell lineage commitment [121, 122].

Notch signaling has also been suggested to regulate HSC renewal (Fig 2b), but this is still highly controversial. Notch1 was shown to be required for generating HSCs during embryonic development [123-125]. In addition, osteoblast-specific expression of parathyroid hormone related protein receptor increased Jagged1 expression, which resulted in HSC expansion, implicating that Jagged1-mediated Notch signaling creates a HSC niche in the bone [126]. Overexpression of either NICD or the Notch downstream target Hes1 resulted in increased self-renewal of HSCs [127, 128]. In contrast, loss-of-function studies using a dominant negative form of the coactivator protein MAML or a conditional RBPjκ knockout in adult HSCs did not show any phenotypes, suggesting that Notch signaling is dispensable for maintaining HSC homeostasis in the adult in vivo [129]. In summary, these studies suggest that γ-secretase inhibition is likely be tolerated in self-renewal of adult HSCs, but will impair T cell development and suppress or alter many hematopoietic lineages in the adult.

Notch signaling in skin barrier defects

The skin is another organ that goes through robust self-renewal throughout life, and dysfunction in the skin barrier can result in dehydration, infection, or cancer. As in the gut, adult stem cells can be found in two places in the skin, a proliferative population in the epidermis and the hair bulb, and a quiescent population in the bulge region of the hair follicles [73]. The epidermis consists of four layers: basal (innermost), spinous, granular, and cornified layers, which express different cellular markers. The adult skin epidermal stem cells reside in the basal layer, and as they detach and migrate outward, they commit to terminal differentiation [130, 131].

Notch signaling regulates differentiation and proliferation of adult stem cells in the epidermis (Fig 2c) [80, 132-134]. Conditional gain- (NICD overexpression) and loss-of-function (Notch1−/− or RBPjκ−/−) of Notch signaling in the epidermis during development and in the adult suggests that canonical Notch signaling contributes to spinous cell differentiation in vivo [135-138]. However, while embryonic ablation of RBPjκ in the epidermis resulted in hypoplasia [135], its postnatal deletion resulted in hyperplasia and increased susceptibility to tumorigenesis [137, 139], suggesting that different mechanisms underlie the proliferative effects of the Notch signaling in the adult stem cells. This led to a surprising proposal that Notch1 functions in the adult skin as a “tumor suppressor”, in contrast to its well known oncogenic functions in many other tissues [139, 140]. However, this view was challenged by a recent study using a multistage chemical skin carcinogenesis paradigm on a chimeric mouse model of Notch deletion in the skin. Rather than Notch functioning as a “tumor suppressor” in the skin, loss of Notch1 was shown to act non-cell autonomously by creating a microenvironment that promotes carcinogenesis[137].

Moderate reduction of Notch signaling in the skin can dose-dependently increase the susceptibility to tumorigenesis. While conditional knockout of either Notch2 or 3 alone in the postnatal epidermis exhibited no phenotype, stepwise deletion of Notch paralogs dose-dependently accelerated skin carcinogenesis, suggesting that Notch1, 2 and 3 were not redundant but additive [137]. Similarly, moderate reduction of γ-secretase activity (either PS1+/−, PS1+/−;PS2−/−, Nicastrin+/−, or Aph1a+/− ) increased the risk of developing squamous cell carcinoma [141, 142]. Another study has shown that skin barrier defects caused by loss of Notch signaling can induce systemic B-lymphoproliferative disorder in newborn mice through a dose-dependent secretion of thymic stromal lymphopoietin (TSLP), which can eventually lead to atopic dermatitis and asthma in adult animals [143, 144]. While TSLP can be useful as a biomarker for Notch-mediated skin barrier defects, these results raise the concern that even a moderate reduction of Notch signaling can increase susceptibility to carcinogenesis in the skin and simultaneously add risks of atopic dermatitis and asthma. Indeed, PS1+/−;PS2−/− mice with reduced dosages of γ-secretase in vivo developed autoimmune phenotypes [142, 145], most likely in synergy with skin barrier defects. Recently, haploinsufficiency of the γ-secretase component genes PS1, Nicastrin and Pen2 have been associated with familial Acne Inversa, highlighting the sensitivity of the skin to reduction in γ-secretase activity [146].

Notch signaling in adult neurogenesis and synaptic plasticity

Unlike the intestine, blood, or skin where massive cell renewal is continuous throughout life, the vast majority of neurogenesis takes place during embryonic development. Adult neurogenesis occurs in two restricted regions of the brain - in the subgranular zone (SGZ) of the hippocampus and the subventicular zone (SVZ) of the lateral ventricle. Although the functional role of adult neurogenesis is not fully understood, it has been implicated in hippocampus-dependent learning and memory, and neurological disorders such as epilepsy and AD. Moreover, adult neurogenesis can be enhanced upon injury, exercise, or an enriched environment [147, 148].

Notch signaling has classically been shown to play a critical role in the maintenance of neural progenitor identity and the inhibition of neuronal differentiation during early development [149]. The current view holds that while Notch signaling may inhibit the neural fate in the early developmental stage, it can also instruct or permissively allow fate choices between different subtypes of neural cells in the later stage, serving as a binary switch of cell fate determination [150]. Emerging evidence also suggests multiple roles for Notch signaling in adult neurogenesis (Fig 2d) [80, 151]. During adult neurogenesis, neural stem cells (NSCs), which specifically express cellular markers such as glial fibrillary acidic protein (GFAP), glutamate-aspartate transporter (GLAST), and Nestin, divide slowly and asymmetrically to produce NSCs and TA cells. TA cells proliferate rapidly, exit from cell cycle and produce neural progenitor cells (NPCs), which migrate from SGZ and SVZ to the granule cell layer or olfactory bulb, respectively, where they mature into neurons. In the SGZ, a study using GFAP-CreERT Notch1 knockout mice and reciprocal experiments employing NICD transgenic mice suggested that Notch signaling promotes proliferation and inhibits cell cycle exit from TA cells to NPCs [152]. Notably, two recent studies using GLAST-CreERT and Nestin-CreERT inducible RBPjκ knockout mice, which completely ablate canonical Notch signaling in the NSCs within the SGZ or SVZ, suggest that Notch signaling instead inhibits entry from NSC pools into TA status and is critical in long-term maintenance of adult NSCs [153, 154]. The discrepancy from the previous study may be due to residual signaling by other Notch receptors in the Notch1 knockout mice. Although forebrain ependymal cells are not formally considered as stem cells, they were shown to give rise to neuroblasts and astrocytes upon recovery from stroke, and Notch signaling plays an active role in maintaining their quiescence [155].

Notch signaling has also been shown to be involved in neurite remodeling and synaptic plasticity in the adult brain [156]. Several studies have indicated that Notch signaling modulates neurite outgrowth in vitro as well as in vivo, although the underlying mechanism still remains unknown (Fig 2d) [152, 157, 158]. A study using hippocampal slices from Notch antisense transgenic mice has suggested that Notch is involved in long-term potentiation [159]. Roles for Notch signaling were also implicated in learning and memory. In Drosophila, Notch signaling was shown to play a role in long-term memory [160, 161]. In mice, postnatal forebrain-specific deletion of PS1, both PS1 and PS2, or Nicastrin resulted in impairment in spatial memory and/or age-related neurodegeneration [162-164], although direct evidence of whether these phenotypes can be attributed to the lack of Notch signaling has not yet been provided. Notch1+/− and RBPjκ+/− mice were both shown to exhibit spatial memory deficits [165]; however, hippocampus-specific and postnatal deletion of Notch signaling will have to be examined to exclude the possibility that it may be secondary to developmental effects caused by the reduction of Notch signaling.

Many questions still remain, such as how much reduction of Notch signaling can be tolerated, is this threshold different in various individuals (a quantitative trait) or how different Notch paralogs contribute to adult neurogenesis, neuritogenesis, or memory and learning. Lastly, the role of adult neurogenesis in AD has been recently discussed in several reviews; however, how Notch signaling may be involved in its pathogenesis is still largely unknown [156, 166-169].

Notch signaling in adult skeletal muscle

Satellite cells are stem cells of mature skeletal muscle that are responsible for its regenerative potential, and dysfunction in this regenerative capacity can lead to sarcopenia (loss of muscle mass). Although the majority of satellite cells are mitotically quiescent, muscles can go through robust regeneration throughout life, upon homeostatic demand, hypertrophy, or need for repair [170].

Loss of Delta1 ligands during muscle development leads to the premature differentiation of muscle progenitors and a concomitant loss of satellite cells, causing severe muscle hypotrophy in newborns [171]. Notch signaling also plays a key role in regulating adult muscle regeneration (Fig 2e) [80, 170, 172-174]. In a myofiber explant culture system that mimics muscle injury, Notch1 was rapidly activated in satellite cells and promoted cell proliferation of myogenic precursor cells that express the premyoblast marker, Pax3 [175]. It was also shown that the Notch antagonist, Numb, was asymmetrically localized in dividing cells, and that attenuation of Notch signaling was required for fate determination toward myoblasts. Furthermore, satellite cells were discovered to consist of a heterogeneous population of stem cells and committed progenitors. Notch3 expression was particularly elevated in the former cell population [176]. It was additionally shown that Notch signaling is important in the early phase of response after injury, and a temporal switch from Notch to Wnt signaling in the later stage of regeneration was critical for progenitor cells to differentiate [177].

Interestingly, modulating Notch activity can restore a decrease in regenerative capacity in aging muscle. In aged muscle, decreased Delta1 expression in satellite cells correlated with an age-dependent decrease in its regenerative capacity [178]. However, this was rescued by increasing Delta1 expression levels, suggesting that regenerative capacity is not intrinsically impaired in older satellite cells. Furthermore, an in vivo study using parabiotic pairing between young and old mice demonstrated that some systemic cues could increase Delta1 expression and activate Notch signaling in aged muscle [179]. One interesting caveat of this study was that this effect was not muscle-specific; hepatocyte proliferation was also enhanced. Wnt3a, TGFβ, and testosterone have emerged as candidate systemic factors although the molecular mechanisms underlying their effects still remain to be elucidated [180, 181].

In summary these studies suggest that the reduction of Notch signaling by γ-secretase inhibition could mimic the age-dependent loss of regenerative capacity in adult muscle. Simultaneous introduction of systemic factors that maintain satellite cell regenerative capacity may be a useful therapeutic strategy to antagonize the undesirable effects of γ-secretase inhibition in adult muscle and possibly other organs. However, further investigation is still needed to identify these factors to be able to precisely manipulate the dose of Notch signaling within the safe therapeutic window.

Notch signaling and human diseases

Given its role in regulating many fundamental processes such as cell proliferation, differentiation and apoptosis in a variety of tissues, the Notch signaling pathway is highly regulated. Accordingly, misregulation or misexpression of Notch pathway components has been linked to cancer (e.g., T-ALL) and adult-onset diseases (e.g., CADASIL and multiple sclerosis). Aberrant Notch signaling has also been associated with several developmental syndromes but these are beyond the scope of this review [182].

Notch and T-ALL

As Notch signaling is required for multiple stages of T-cell development, it is not surprising that the human Notch1 gene was discovered for its involvement in T-ALL, an aggressive blood cancer resulting from malignant transformation of T-cell precursors [27, 183-187]. A t(7;9) chromosomal translocation in a small subset (~1%) of T-ALL cases leads to expression of an extracellular domain-truncated, constitutively active form of Notch1 [183]. A more general role for Notch signaling in human T-ALL was recognized when activating Notch1 mutations were subsequently found in >50% of T-ALL cases [27]. These activating Notch1 mutations are clustered in two regions, the extracellular heterodimerization domain (HD) and the C-terminal PEST domain. The HD mutations found in T-ALL either destabilize Notch heterodimers or displace the S2 site to the outside of NRR, thereby leading to ligand-independent Notch signaling [5, 188, 189]. Partial or complete loss of the PEST domain increases NICD half-life, thus enhancing Notch signaling [189, 190]. Notably, mutations in HD and the PEST domain were also found in cis in the same NOTCH1 allele; reporter gene assays confirmed the synergistic effect of these two groups of mutations [27]. Recently, Sulis et al found in a subset of T-ALLs a novel type of mutation resulting in extracellular juxtamembrane expansions (JMEs) [191]. These mutations markedly enhance Notch signaling in a ligand-independent manner, but do not affect the stability of Notch heterodimers and still require prior S2 cleavage [191]. It is still unclear how JME mutations affect S2 site cleavage.

While the association of activating Notch mutations with T-ALL is clear, the downstream targets of Notch signaling and how they relate to the pathogenesis are far from being completely understood. A large body of evidence indicates that the oncogenic transcription factor Myc is a direct Notch target in T-ALL [192-194]. Recently, accumulating evidence also suggests that Notch signaling induces cell transformation via multiple signal pathways, including NF-κB [195], PI3K–AKT [196], and the NFAT signaling pathways [197].

The discovery of activating Notch mutations in more than 50% of cases of human T-ALL provides a rationale for Notch as a therapeutic target. Indeed, GSIs were proposed as potential drugs for T-ALL [27, 93]. However, the application of GSIs in T-ALL has been hampered by the limited anti-leukemic effects of GSIs [27] and severe gastrointestinal toxicity due to the inhibition of Notch signaling in the gut, as discussed earlier [90, 91, 93]. Still, GSIs appear to sensitize T-ALL cells to chemotherapy [192, 198] and a recent report showed that the combination therapy of GSIs and glucocorticoids improved the anti-leukemic effects and reduced GSI-induced intestinal toxicity [184]. It was suggested that inhibition of Notch signaling by GSIs leads to upregulation of glucocorticoid receptor expression presumably via the transcriptional repressor HES1, whereas glucocorticoid treatment diminishes GSI-induced intestinal toxicity by upregulation of Ccnd2 [94, 184]. Although the molecular mechanisms involved remain to be elucidated, the combination therapy of GSIs and glucocorticoids may provide a promising strategy to target T-ALL.

Notch and CADASIL

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common form of hereditary adult-onset stroke and vascular dementia that is caused by mutations in Notch3 [199-201]. The Notch3 receptor is required for arterial identity and maturation of vascular smooth muscle cells (vSMCs) [202] and Notch3 is predominantly expressed in vSMCs in adults [203]. The key pathological features of CADASIL include the loss of vSMCs and the accumulation of Notch3 extracellular domains (ECDs) in the plasma membranes of vSMCs [200, 203]. To date, more than 170 mutations in Notch3 have been identified to be linked with CADASIL from different pedigrees [204]. These mutations span exons 2-24, with most clustering in exons 3 and 4, which encode EGF repeats 2-5. Most are missense mutations and in some cases short in-frame deletions. Strikingly, almost all CADASIL-causing mutations hitherto described result in loss or gain of 1, or rarely 3, cysteines in 1 of the 34 EGF repeats in Notch3 [200]. Normally, each EGF repeat contains 6 cysteines, whereas all Notch3 mutations in CADASIL lead to an odd number of cysteines in the affected repeat.

Although the association of Notch3 mutations with CADASIL has been well-established, the underlying molecular mechanisms remain unclear. Similar to AD, both loss- and gain-of-function models have been hypothesized for CADASIL [205, 206]. It is possible that the unpaired cysteines may lead to a misfolded Notch3 ECD, which may compromise Notch3 signaling by affecting trafficking, post-translational modifications, S1 cleavage and/or ligand binding [207], and ultimately affecting vSMC maturation and small artery function. However, in vitro downstream reporter gene assays show no significant differences between wild-type and most CADASIL-associated mutant Notch3 receptors [208, 209]. The only exceptions are mutations that map to the ligand-binding domain, which lead to decreased activity in reporter gene assays and are associated with an earlier onset of the disease [210]. In addition, although a complete loss of Notch3 leads to structural and functional alterations in small arteries as well as increased susceptibility to stroke in mice, it does not lead to CADASIL-like symptoms [202, 211, 212]. Therefore, a loss of Notch3 function alone cannot cause this disease. Consistent with this idea, disease-associated mutant Notch3 receptors are functional in vivo and can rescue the arterial defects in Notch3 knockout mice [213]. Alternatively, mutant Notch3 with unpaired cysteines may gain some novel disease-causing functions. Several lines of evidence favor this gain-of-function model. For instance, compared to wild-type Notch3, CADASIL-causing mutants are more prone to aggregation [214] and accumulate in the membranes of vSMCs [203]. Additionally, CADASIL-associated Notch3 mutants can accumulate in the endoplasmic reticulum and impair cell proliferation [215]. Interestingly, transgenic mice that express higher levels of mutant Notch3 (above a certain threshold) develop several main pathological features of CADASIL [216]. Therefore, the current data favor a neomorphic effect of CADASIL mutations. How Notch3 ECD mutations and protein aggregation specifically alter vSMC function and lead to the disease still need to be elucidated. It is unclear whether they act by affecting normal Notch signaling processes and/or other cellular pathways. Regardless, these studies have revealed a role for normal Notch signaling in the maintenance of vSMCs and small artery functions.

Notch and Multiple sclerosis

The Notch signaling pathway has also been implicated in multiple sclerosis (MS), a central nervous system (CNS) disorder characterized by chronic inflammation, demyelination of axons as well as damage to axons and oligodendrocytes [207, 217-219]. A key feature of MS is the impaired differentiation of oligodendrocyte precursor cells (OPCs) into myelin-forming oligodendrocytes [220].

Canonical Notch signaling prevents oligodendrocyte maturation and myelin formation by blocking OPC differentiation in the developing CNS [221, 222]. It was therefore speculated that canonical Notch signaling might be activated in MS lesions to prevent OPCs from differentiating into myelin-forming oligodendrocytes [223]. Consistent with this idea, Notch1, Jagged1 and the downstream target Hes5 are expressed in active MS lesions [224]. Other studies show that Notch1 and Jagged1 ligand are also expressed in remyelinated lesions [225, 226] although the functional significance is unclear. Notably, recent in vivo data showed that Olig1-Cre-mediated loss of Notch1 in OPCs led to accelerated OPC differentiation and remyelination in demyelinating lesions [227], indicating a role for Notch signaling in preventing OPC differentiation in MS lesions.

Non-canonical Notch signaling pathways are also involved in MS. Under physiological conditions, Notch is activated by the non-canonical ligand F3/contactin to promote OPC differentiation and upregulate myelin-related protein MAG [11, 228]. Interestingly, during the repair of MS lesions, while F3/contactin activates Notch signaling in OPCs and releases NICD via γ-secretase cleavage, the NICD molecule is trapped in the cytoplasm, rather than translocating into the nucleus, which is required for myelinogenesis [229]. Normally, NICD nuclear translocation is mediated by the nuclear transporter importin-β [230]. However, in demyelinated lesions, an inhibitor of nuclear importin-β, TAT-interacting protein TIP30, is markedly upregulated and forms aggregates with nuclear importin-β, thereby blocking NICD nuclear translocation [229].

Collectively, both canonical and non-canonical Notch signaling pathways appear to be involved in the pathogenesis of MS. It should be noted that unlike in T-ALL and CADASIL, where a major role for Notch signaling is well established, the Notch pathway might be just one of several signaling pathways to be involved in MS [220, 231]. Interestingly, pharmacological inhibition of Notch signaling with GSIs had beneficial effects in the mouse experimental autoimmune encephalomyelitis model of MS and therefore, warrants further study [232-234].

Conclusions and Perspectives

Notch signaling plays a critical role in various tissues and is used reiteratively at all stages of development, and during adult tissue renewal. It is clear that different functional thresholds of Notch signaling exist for various contexts. The central role of proteolysis (by both ADAMs and γ-secretase) in the Notch signaling mechanism underlies the major concern towards the use of both α-secretase agonists and GSIs in the chronic treatment of AD. On the other hand, GSIs have emerged as possible drugs for treatment of several types of cancers involving Notch. Regardless of clinical application, the studies summarized here provide a framework for organ systems that might be especially sensitive to chronic reduction or enhancement in Notch activity. Paralog-specific functions will have to also be addressed in future studies. Even strategies that avoid simple γ-secretase inhibition (e.g., γ-secretase modulators, BACE inhibitors, targeting specific γ-secretase complexes) will need to be extensively characterized in various self-renewing systems. Combination therapies could be promising.

Acknowledgements

We are especially grateful to Dr. Raphael Kopan for his critical reading of this review. We also thank our colleagues, Shuang Chen, and Drs. Scott Boyle, Shawn Demehri, Matt Hass, Zhenyi Liu and Mitsuru Morimoto for comments and discussion. Our research on the mechanisms and functions of Notch signaling and γ-secretase in the Kopan Lab is supported by JSPS Postdoctoral Fellowships for Research Abroad to C. Sato and National Institutes of Health grants GM55479 and AG025973 to R. Kopan and ADRC P50 AG05681 to J. Morris.

References

  • 1.Artavanis-Tsakonas S, Rand MD, Lake RJ. Notch signaling: cell fate control and signal integration in development. Science. 1999;284(5415):770–6. doi: 10.1126/science.284.5415.770. [DOI] [PubMed] [Google Scholar]
  • 2.Bray SJ. Notch signalling: a simple pathway becomes complex. Nat Rev Mol Cell Biol. 2006;7(9):678–89. doi: 10.1038/nrm2009. [DOI] [PubMed] [Google Scholar]
  • 3.Fortini ME. Notch signaling: the core pathway and its posttranslational regulation. Dev Cell. 2009;16(5):633–47. doi: 10.1016/j.devcel.2009.03.010. [DOI] [PubMed] [Google Scholar]
  • 4.Lai EC. Notch signaling: control of cell communication and cell fate. Development. 2004;131(5):965–73. doi: 10.1242/dev.01074. [DOI] [PubMed] [Google Scholar]
  • 5.Gordon WR, Roy M, Vardar-Ulu D, Garfinkel M, Mansour MR, Aster JC, et al. Structure of the Notch1-negative regulatory region: implications for normal activation and pathogenic signaling in T-ALL. Blood. 2009;113(18):4381–90. doi: 10.1182/blood-2008-08-174748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sanchez-Irizarry C, Carpenter AC, Weng AP, Pear WS, Aster JC, Blacklow SC. Notch subunit heterodimerization and prevention of ligand-independent proteolytic activation depend, respectively, on a novel domain and the LNR repeats. Mol Cell Biol. 2004;24(21):9265–73. doi: 10.1128/MCB.24.21.9265-9273.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Gordon WR, Arnett KL, Blacklow SC. The molecular logic of Notch signaling--a structural and biochemical perspective. J Cell Sci. 2008;121(Pt 19):3109–19. doi: 10.1242/jcs.035683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Gordon WR, Vardar-Ulu D, Histen G, Sanchez-Irizarry C, Aster JC, Blacklow SC. Structural basis for autoinhibition of Notch. Nat Struct Mol Biol. 2007;14(4):295–300. doi: 10.1038/nsmb1227. [DOI] [PubMed] [Google Scholar]
  • 9.Kopan R, Ilagan MX. The canonical Notch signaling pathway: unfolding the activation mechanism. Cell. 2009;137(2):216–33. doi: 10.1016/j.cell.2009.03.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Gordon WR, Vardar-Ulu D, L'Heureux S, Ashworth T, Malecki MJ, Sanchez-Irizarry C, et al. Effects of S1 cleavage on the structure, surface export, and signaling activity of human Notch1 and Notch2. PLoS One. 2009;4(8):e6613. doi: 10.1371/journal.pone.0006613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.D'Souza B, Miyamoto A, Weinmaster G. The many facets of Notch ligands. Oncogene. 2008;27(38):5148–67. doi: 10.1038/onc.2008.229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Tien AC, Rajan A, Bellen HJ. A Notch updated. J Cell Biol. 2009;184(5):621–9. doi: 10.1083/jcb.200811141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wang W, Struhl G. Distinct roles for Mind bomb, Neuralized and Epsin in mediating DSL endocytosis and signaling in Drosophila. Development. 2005;132(12):2883–94. doi: 10.1242/dev.01860. [DOI] [PubMed] [Google Scholar]
  • 14.Le Borgne R. Regulation of Notch signalling by endocytosis and endosomal sorting. Curr Opin Cell Biol. 2006;18(2):213–22. doi: 10.1016/j.ceb.2006.02.011. [DOI] [PubMed] [Google Scholar]
  • 15.Fortini ME, Bilder D. Endocytic regulation of Notch signaling. Curr Opin Genet Dev. 2009;19(4):323–8. doi: 10.1016/j.gde.2009.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Parks AL, Klueg KM, Stout JR, Muskavitch MA. Ligand endocytosis drives receptor dissociation and activation in the Notch pathway. Development. 2000;127(7):1373–85. doi: 10.1242/dev.127.7.1373. [DOI] [PubMed] [Google Scholar]
  • 17.Nichols JT, Miyamoto A, Olsen SL, D'Souza B, Yao C, Weinmaster G. DSL ligand endocytosis physically dissociates Notch1 heterodimers before activating proteolysis can occur. J Cell Biol. 2007;176(4):445–58. doi: 10.1083/jcb.200609014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bozkulak EC, Weinmaster G. Selective use of ADAM10 and ADAM17 in activation of Notch1 signaling. Mol Cell Biol. 2009;29(21):5679–95. doi: 10.1128/MCB.00406-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.van Tetering G, van Diest P, Verlaan I, van der Wall E, Kopan R, Vooijs M. Metalloprotease ADAM10 is required for Notch1 site 2 cleavage. J Biol Chem. 2009;284(45):31018–27. doi: 10.1074/jbc.M109.006775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Deuss M, Reiss K, Hartmann D. Part-time alpha-secretases: the functional biology of ADAM 9, 10 and 17. Curr Alzheimer Res. 2008;5(2):187–201. doi: 10.2174/156720508783954686. [DOI] [PubMed] [Google Scholar]
  • 21.Edwards DR, Handsley MM, Pennington CJ. The ADAM metalloproteinases. Mol Aspects Med. 2008;29(5):258–89. doi: 10.1016/j.mam.2008.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Reiss K, Saftig P. The “a disintegrin and metalloprotease” (ADAM) family of sheddases: physiological and cellular functions. Semin Cell Dev Biol. 2009;20(2):126–37. doi: 10.1016/j.semcdb.2008.11.002. [DOI] [PubMed] [Google Scholar]
  • 23.Delwig A, Rand MD. Kuz and TACE can activate Notch independent of ligand. Cell Mol Life Sci. 2008;65(14):2232–43. doi: 10.1007/s00018-008-8127-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sawey ET, Crawford HC. Metalloproteinases and cell fate: Notch just ADAMs anymore. Cell Cycle. 2008;7(5):566–9. doi: 10.4161/cc.7.5.5531. [DOI] [PubMed] [Google Scholar]
  • 25.Rand MD, Grimm LM, Artavanis-Tsakonas S, Patriub V, Blacklow SC, Sklar J, et al. Calcium depletion dissociates and activates heterodimeric notch receptors. Mol Cell Biol. 2000;20(5):1825–35. doi: 10.1128/mcb.20.5.1825-1835.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mumm JS, Schroeter EH, Saxena MT, Griesemer A, Tian X, Pan DJ, et al. A ligand-induced extracellular cleavage regulates gamma-secretase-like proteolytic activation of Notch1. Mol Cell. 2000;5(2):197–206. doi: 10.1016/s1097-2765(00)80416-5. [DOI] [PubMed] [Google Scholar]
  • 27.Weng AP, Ferrando AA, Lee W, Morris JPt, Silverman LB, Sanchez-Irizarry C, et al. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science. 2004;306(5694):269–71. doi: 10.1126/science.1102160. [DOI] [PubMed] [Google Scholar]
  • 28.Li H, Wolfe MS, Selkoe DJ. Toward structural elucidation of the gamma-secretase complex. Structure. 2009;17(3):326–34. doi: 10.1016/j.str.2009.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bergmans BA, De Strooper B. gamma-secretases: from cell biology to therapeutic strategies. Lancet Neurol. 2010;9(2):215–26. doi: 10.1016/S1474-4422(09)70332-1. [DOI] [PubMed] [Google Scholar]
  • 30.Okochi M, Steiner H, Fukumori A, Tanii H, Tomita T, Tanaka T, et al. Presenilins mediate a dual intramembranous gamma-secretase cleavage of Notch-1. EMBO J. 2002;21(20):5408–16. doi: 10.1093/emboj/cdf541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Seugnet L, Simpson P, Haenlin M. Requirement for dynamin during Notch signaling in Drosophila neurogenesis. Dev Biol. 1997;192(2):585–98. doi: 10.1006/dbio.1997.8723. [DOI] [PubMed] [Google Scholar]
  • 32.Windler SL, Bilder D. Endocytic internalization routes required for delta/notch signaling. Curr Biol. 2010;20(6):538–43. doi: 10.1016/j.cub.2010.01.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Vaccari T, Lu H, Kanwar R, Fortini ME, Bilder D. Endosomal entry regulates Notch receptor activation in Drosophila melanogaster. J Cell Biol. 2008;180(4):755–62. doi: 10.1083/jcb.200708127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Tagami S, Okochi M, Yanagida K, Ikuta A, Fukumori A, Matsumoto N, et al. Regulation of Notch signaling by dynamic changes in the precision of S3 cleavage of Notch-1. Mol Cell Biol. 2008;28(1):165–76. doi: 10.1128/MCB.00863-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Barrick D, Kopan R. The Notch transcription activation complex makes its move. Cell. 2006;124(5):883–5. doi: 10.1016/j.cell.2006.02.028. [DOI] [PubMed] [Google Scholar]
  • 36.Kovall RA. More complicated than it looks: assembly of Notch pathway transcription complexes. Oncogene. 2008;27(38):5099–109. doi: 10.1038/onc.2008.223. [DOI] [PubMed] [Google Scholar]
  • 37.Zolkiewska A. ADAM proteases: ligand processing and modulation of the Notch pathway. Cell Mol Life Sci. 2008;65(13):2056–68. doi: 10.1007/s00018-008-7586-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bland CE, Kimberly P, Rand MD. Notch-induced proteolysis and nuclear localization of the Delta ligand. J Biol Chem. 2003;278(16):13607–10. doi: 10.1074/jbc.C300016200. [DOI] [PubMed] [Google Scholar]
  • 39.LaVoie MJ, Selkoe DJ. The Notch ligands, Jagged and Delta, are sequentially processed by alpha-secretase and presenilin/gamma-secretase and release signaling fragments. J Biol Chem. 2003;278(36):34427–37. doi: 10.1074/jbc.M302659200. [DOI] [PubMed] [Google Scholar]
  • 40.Sapir A, Assa-Kunik E, Tsruya R, Schejter E, Shilo BZ. Unidirectional Notch signaling depends on continuous cleavage of Delta. Development. 2005;132(1):123–32. doi: 10.1242/dev.01546. [DOI] [PubMed] [Google Scholar]
  • 41.Mishra-Gorur K, Rand MD, Perez-Villamil B, Artavanis-Tsakonas S. Down-regulation of Delta by proteolytic processing. J Cell Biol. 2002;159(2):313–24. doi: 10.1083/jcb.200203117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Qi H, Rand MD, Wu X, Sestan N, Wang W, Rakic P, et al. Processing of the notch ligand delta by the metalloprotease Kuzbanian. Science. 1999;283(5398):91–4. doi: 10.1126/science.283.5398.91. [DOI] [PubMed] [Google Scholar]
  • 43.Dyczynska E, Sun D, Yi H, Sehara-Fujisawa A, Blobel CP, Zolkiewska A. Proteolytic processing of delta-like 1 by ADAM proteases. J Biol Chem. 2007;282(1):436–44. doi: 10.1074/jbc.M605451200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Gravano DM, Manilay JO. Inhibition of proteolysis of Delta-like-1 does not promote or reduce T-cell developmental potential. Immunol Cell Biol. 2010 doi: 10.1038/icb.2010.30. [DOI] [PubMed] [Google Scholar]
  • 45.Muraguchi T, Takegami Y, Ohtsuka T, Kitajima S, Chandana EP, Omura A, et al. RECK modulates Notch signaling during cortical neurogenesis by regulating ADAM10 activity. Nat Neurosci. 2007;10(7):838–45. doi: 10.1038/nn1922. [DOI] [PubMed] [Google Scholar]
  • 46.Sun D, Li H, Zolkiewska A. The role of Delta-like 1 shedding in muscle cell self-renewal and differentiation. J Cell Sci. 2008;121(Pt 22):3815–23. doi: 10.1242/jcs.035493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hass MR, Sato C, Kopan R, Zhao G. Presenilin: RIP and beyond. Semin Cell Dev Biol. 2009;20(2):201–10. doi: 10.1016/j.semcdb.2008.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kopan R, Ilagan MX. Gamma-secretase: proteasome of the membrane? Nat Rev Mol Cell Biol. 2004;5(6):499–504. doi: 10.1038/nrm1406. [DOI] [PubMed] [Google Scholar]
  • 49.Hiratochi M, Nagase H, Kuramochi Y, Koh CS, Ohkawara T, Nakayama K. The Delta intracellular domain mediates TGF-beta/Activin signaling through binding to Smads and has an important bi-directional function in the Notch-Delta signaling pathway. Nucleic Acids Res. 2007;35(3):912–22. doi: 10.1093/nar/gkl1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chung HM, Struhl G. Nicastrin is required for Presenilin-mediated transmembrane cleavage in Drosophila. Nat Cell Biol. 2001;3(12):1129–32. doi: 10.1038/ncb1201-1129. [DOI] [PubMed] [Google Scholar]
  • 51.Lopez-Schier H, St Johnston D. Drosophila nicastrin is essential for the intramembranous cleavage of notch. Dev Cell. 2002;2(1):79–89. doi: 10.1016/s1534-5807(01)00109-5. [DOI] [PubMed] [Google Scholar]
  • 52.Aster JC, Pear WS, Blacklow SC. Notch signaling in leukemia. Annu Rev Pathol. 2008;3(587-613) doi: 10.1146/annurev.pathmechdis.3.121806.154300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Haines N, Irvine KD. Glycosylation regulates Notch signalling. Nat Rev Mol Cell Biol. 2003;4(10):786–97. doi: 10.1038/nrm1228. [DOI] [PubMed] [Google Scholar]
  • 54.Ilagan MX, Kopan R. SnapShot: notch signaling pathway. Cell. 2007;128(6):1246. doi: 10.1016/j.cell.2007.03.011. [DOI] [PubMed] [Google Scholar]
  • 55.Nichols JT, Miyamoto A, Weinmaster G. Notch signaling--constantly on the move. Traffic. 2007;8(8):959–69. doi: 10.1111/j.1600-0854.2007.00592.x. [DOI] [PubMed] [Google Scholar]
  • 56.Stanley P. Regulation of Notch signaling by glycosylation. Curr Opin Struct Biol. 2007;17(5):530–5. doi: 10.1016/j.sbi.2007.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Takeuchi H, Haltiwanger RS. Role of glycosylation of Notch in development. Semin Cell Dev Biol. 2010 doi: 10.1016/j.semcdb.2010.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Swiatek PJ, Lindsell CE, del Amo FF, Weinmaster G, Gridley T. Notch1 is essential for postimplantation development in mice. Genes Dev. 1994;8(6):707–19. doi: 10.1101/gad.8.6.707. [DOI] [PubMed] [Google Scholar]
  • 59.Conlon RA, Reaume AG, Rossant J. Notch1 is required for the coordinate segmentation of somites. Development. 1995;121(5):1533–45. doi: 10.1242/dev.121.5.1533. [DOI] [PubMed] [Google Scholar]
  • 60.Krebs LT, Xue Y, Norton CR, Shutter JR, Maguire M, Sundberg JP, et al. Notch signaling is essential for vascular morphogenesis in mice. Genes Dev. 2000;14(11):1343–52. [PMC free article] [PubMed] [Google Scholar]
  • 61.Hamada Y, Kadokawa Y, Okabe M, Ikawa M, Coleman JR, Tsujimoto Y. Mutation in ankyrin repeats of the mouse Notch2 gene induces early embryonic lethality. Development. 1999;126(15):3415–24. doi: 10.1242/dev.126.15.3415. [DOI] [PubMed] [Google Scholar]
  • 62.Huppert SS, Le A, Schroeter EH, Mumm JS, Saxena MT, Milner LA, et al. Embryonic lethality in mice homozygous for a processing-deficient allele of Notch1. Nature. 2000;405(6789):966–70. doi: 10.1038/35016111. [DOI] [PubMed] [Google Scholar]
  • 63.Oka C, Nakano T, Wakeham A, de la Pompa JL, Mori C, Sakai T, et al. Disruption of the mouse RBP-J kappa gene results in early embryonic death. Development. 1995;121(10):3291–301. doi: 10.1242/dev.121.10.3291. [DOI] [PubMed] [Google Scholar]
  • 64.Shi S, Stahl M, Lu L, Stanley P. Canonical Notch signaling is dispensable for early cell fate specifications in mammals. Mol Cell Biol. 2005;25(21):9503–8. doi: 10.1128/MCB.25.21.9503-9508.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Wong PC, Zheng H, Chen H, Becher MW, Sirinathsinghji DJ, Trumbauer ME, et al. Presenilin 1 is required for Notch1 and DII1 expression in the paraxial mesoderm. Nature. 1997;387(6630):288–92. doi: 10.1038/387288a0. [DOI] [PubMed] [Google Scholar]
  • 66.Shen J, Bronson RT, Chen DF, Xia W, Selkoe DJ, Tonegawa S. Skeletal and CNS defects in Presenilin-1-deficient mice. Cell. 1997;89(4):629–39. doi: 10.1016/s0092-8674(00)80244-5. [DOI] [PubMed] [Google Scholar]
  • 67.Herreman A, Hartmann D, Annaert W, Saftig P, Craessaerts K, Serneels L, et al. Presenilin 2 deficiency causes a mild pulmonary phenotype and no changes in amyloid precursor protein processing but enhances the embryonic lethal phenotype of presenilin 1 deficiency. Proc Natl Acad Sci U S A. 1999;96(21):11872–7. doi: 10.1073/pnas.96.21.11872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Li T, Ma G, Cai H, Price DL, Wong PC. Nicastrin is required for assembly of presenilin/gamma-secretase complexes to mediate Notch signaling and for processing and trafficking of beta-amyloid precursor protein in mammals. J Neurosci. 2003;23(8):3272–7. doi: 10.1523/JNEUROSCI.23-08-03272.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ma G, Li T, Price DL, Wong PC. APH-1a is the principal mammalian APH-1 isoform present in gamma-secretase complexes during embryonic development. J Neurosci. 2005;25(1):192–8. doi: 10.1523/JNEUROSCI.3814-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Nguyen V, Hawkins C, Bergeron C, Supala A, Huang J, Westaway D, et al. Loss of nicastrin elicits an apoptotic phenotype in mouse embryos. Brain Res. 2006;1086(1):76–84. doi: 10.1016/j.brainres.2006.02.122. [DOI] [PubMed] [Google Scholar]
  • 71.Serneels L, Dejaegere T, Craessaerts K, Horre K, Jorissen E, Tousseyn T, et al. Differential contribution of the three Aph1 genes to gamma-secretase activity in vivo. Proc Natl Acad Sci U S A. 2005;102(5):1719–24. doi: 10.1073/pnas.0408901102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Serneels L, Van Biervliet J, Craessaerts K, Dejaegere T, Horre K, Van Houtvin T, et al. gamma-Secretase heterogeneity in the Aph1 subunit: relevance for Alzheimer's disease. Science. 2009;324(5927):639–42. doi: 10.1126/science.1171176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Li L, Clevers H. Coexistence of quiescent and active adult stem cells in mammals. Science. 2010;327(5965):542–5. doi: 10.1126/science.1180794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Lopez-Garcia C, Klein AM, Simons BD, Winton DJ. Intestinal stem cell replacement follows a pattern of neutral drift. Science. 2010;330(6005):822–5. doi: 10.1126/science.1196236. [DOI] [PubMed] [Google Scholar]
  • 75.Snippert HJ, van der Flier LG, Sato T, van Es JH, van den Born M, Kroon-Veenboer C, et al. Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell. 2010;143(1):134–44. doi: 10.1016/j.cell.2010.09.016. [DOI] [PubMed] [Google Scholar]
  • 76.Barker N, van de Wetering M, Clevers H. The intestinal stem cell. Genes Dev. 2008;22(14):1856–64. doi: 10.1101/gad.1674008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Casali A, Batlle E. Intestinal stem cells in mammals and Drosophila. Cell Stem Cell. 2009;4(2):124–7. doi: 10.1016/j.stem.2009.01.009. [DOI] [PubMed] [Google Scholar]
  • 78.Crosnier C, Stamataki D, Lewis J. Organizing cell renewal in the intestine: stem cells, signals and combinatorial control. Nat Rev Genet. 2006;7(5):349–59. doi: 10.1038/nrg1840. [DOI] [PubMed] [Google Scholar]
  • 79.Wang P, Hou SX. Regulation of intestinal stem cells in mammals and Drosophila. J Cell Physiol. 2010;222(1):33–7. doi: 10.1002/jcp.21928. [DOI] [PubMed] [Google Scholar]
  • 80.Liu J, Sato C, Cerletti M, Wagers A. Notch signaling in the regulation of stem cell self-renewal and differentiation. Curr Top Dev Biol. 2010;92(367-409) doi: 10.1016/S0070-2153(10)92012-7. [DOI] [PubMed] [Google Scholar]
  • 81.van der Flier LG, Clevers H. Stem cells, self-renewal, and differentiation in the intestinal epithelium. Annu Rev Physiol. 2009;71(241-60) doi: 10.1146/annurev.physiol.010908.163145. [DOI] [PubMed] [Google Scholar]
  • 82.Chiba S. Notch signaling in stem cell systems. Stem Cells. 2006;24(11):2437–47. doi: 10.1634/stemcells.2005-0661. [DOI] [PubMed] [Google Scholar]
  • 83.Scoville DH, Sato T, He XC, Li L. Current view: intestinal stem cells and signaling. Gastroenterology. 2008;134(3):849–64. doi: 10.1053/j.gastro.2008.01.079. [DOI] [PubMed] [Google Scholar]
  • 84.Riccio O, van Gijn ME, Bezdek AC, Pellegrinet L, van Es JH, Zimber-Strobl U, et al. Loss of intestinal crypt progenitor cells owing to inactivation of both Notch1 and Notch2 is accompanied by derepression of CDK inhibitors p27Kip1 and p57Kip2. EMBO Rep. 2008;9(4):377–83. doi: 10.1038/embor.2008.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.van Es JH, van Gijn ME, Riccio O, van den Born M, Vooijs M, Begthel H, et al. Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature. 2005;435(7044):959–63. doi: 10.1038/nature03659. [DOI] [PubMed] [Google Scholar]
  • 86.Fre S, Huyghe M, Mourikis P, Robine S, Louvard D, Artavanis-Tsakonas S. Notch signals control the fate of immature progenitor cells in the intestine. Nature. 2005;435(7044):964–8. doi: 10.1038/nature03589. [DOI] [PubMed] [Google Scholar]
  • 87.Fre S, Pallavi SK, Huyghe M, Lae M, Janssen KP, Robine S, et al. Notch and Wnt signals cooperatively control cell proliferation and tumorigenesis in the intestine. Proc Natl Acad Sci U S A. 2009;106(15):6309–14. doi: 10.1073/pnas.0900427106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Crosnier C, Vargesson N, Gschmeissner S, Ariza-McNaughton L, Morrison A, Lewis J. Delta-Notch signalling controls commitment to a secretory fate in the zebrafish intestine. Development. 2005;132(5):1093–104. doi: 10.1242/dev.01644. [DOI] [PubMed] [Google Scholar]
  • 89.Vooijs M, Ong CT, Hadland B, Huppert S, Liu Z, Korving J, et al. Mapping the consequence of Notch1 proteolysis in vivo with NIP-CRE. Development. 2007;134(3):535–44. doi: 10.1242/dev.02733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.van Es JH, Clevers H. Notch and Wnt inhibitors as potential new drugs for intestinal neoplastic disease. Trends Mol Med. 2005;11(11):496–502. doi: 10.1016/j.molmed.2005.09.008. [DOI] [PubMed] [Google Scholar]
  • 91.Milano J, McKay J, Dagenais C, Foster-Brown L, Pognan F, Gadient R, et al. Modulation of notch processing by gamma-secretase inhibitors causes intestinal goblet cell metaplasia and induction of genes known to specify gut secretory lineage differentiation. Toxicol Sci. 2004;82(1):341–58. doi: 10.1093/toxsci/kfh254. [DOI] [PubMed] [Google Scholar]
  • 92.Searfoss GH, Jordan WH, Calligaro DO, Galbreath EJ, Schirtzinger LM, Berridge BR, et al. Adipsin, a biomarker of gastrointestinal toxicity mediated by a functional gamma-secretase inhibitor. J Biol Chem. 2003;278(46):46107–16. doi: 10.1074/jbc.M307757200. [DOI] [PubMed] [Google Scholar]
  • 93.Wong GT, Manfra D, Poulet FM, Zhang Q, Josien H, Bara T, et al. Chronic treatment with the gamma-secretase inhibitor LY-411,575 inhibits beta-amyloid peptide production and alters lymphopoiesis and intestinal cell differentiation. J Biol Chem. 2004;279(13):12876–82. doi: 10.1074/jbc.M311652200. [DOI] [PubMed] [Google Scholar]
  • 94.Real PJ, Tosello V, Palomero T, Castillo M, Hernando E, de Stanchina E, et al. Gamma-secretase inhibitors reverse glucocorticoid resistance in T cell acute lymphoblastic leukemia. Nat Med. 2009;15(1):50–8. doi: 10.1038/nm.1900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Cumano A, Godin I. Ontogeny of the hematopoietic system. Annu Rev Immunol. 2007;25(745-85) doi: 10.1146/annurev.immunol.25.022106.141538. [DOI] [PubMed] [Google Scholar]
  • 96.Godin I, Cumano A. The hare and the tortoise: an embryonic haematopoietic race. Nat Rev Immunol. 2002;2(8):593–604. doi: 10.1038/nri857. [DOI] [PubMed] [Google Scholar]
  • 97.Radtke F, Fasnacht N, Macdonald HR. Notch signaling in the immune system. Immunity. 32(1):14–27. doi: 10.1016/j.immuni.2010.01.004. [DOI] [PubMed] [Google Scholar]
  • 98.Sandy AR, Maillard I. Notch signaling in the hematopoietic system. Expert Opin Biol Ther. 2009;9(11):1383–98. doi: 10.1517/14712590903260777. [DOI] [PubMed] [Google Scholar]
  • 99.Yuan JS, Kousis PC, Suliman S, Visan I, Guidos CJ. Functions of notch signaling in the immune system: consensus and controversies. Annu Rev Immunol. 28(343-65) doi: 10.1146/annurev.immunol.021908.132719. [DOI] [PubMed] [Google Scholar]
  • 100.Saito T, Chiba S, Ichikawa M, Kunisato A, Asai T, Shimizu K, et al. Notch2 is preferentially expressed in mature B cells and indispensable for marginal zone B lineage development. Immunity. 2003;18(5):675–85. doi: 10.1016/s1074-7613(03)00111-0. [DOI] [PubMed] [Google Scholar]
  • 101.Tanigaki K, Han H, Yamamoto N, Tashiro K, Ikegawa M, Kuroda K, et al. Notch-RBP-J signaling is involved in cell fate determination of marginal zone B cells. Nat Immunol. 2002;3(5):443–50. doi: 10.1038/ni793. [DOI] [PubMed] [Google Scholar]
  • 102.Maekawa Y, Minato Y, Ishifune C, Kurihara T, Kitamura A, Kojima H, et al. Notch2 integrates signaling by the transcription factors RBP-J and CREB1 to promote T cell cytotoxicity. Nat Immunol. 2008;9(10):1140–7. doi: 10.1038/ni.1649. [DOI] [PubMed] [Google Scholar]
  • 103.Cho OH, Shin HM, Miele L, Golde TE, Fauq A, Minter LM, et al. Notch regulates cytolytic effector function in CD8+ T cells. J Immunol. 2009;182(6):3380–9. doi: 10.4049/jimmunol.0802598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Amsen D, Antov A, Flavell RA. The different faces of Notch in T-helper-cell differentiation. Nat Rev Immunol. 2009;9(2):116–24. doi: 10.1038/nri2488. [DOI] [PubMed] [Google Scholar]
  • 105.Mercher T, Cornejo MG, Sears C, Kindler T, Moore SA, Maillard I, et al. Notch signaling specifies megakaryocyte development from hematopoietic stem cells. Cell Stem Cell. 2008;3(3):314–26. doi: 10.1016/j.stem.2008.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Taghon T, Yui MA, Rothenberg EV. Mast cell lineage diversion of T lineage precursors by the essential T cell transcription factor GATA-3. Nat Immunol. 2007;8(8):845–55. doi: 10.1038/ni1486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Kijima M, Yamaguchi T, Ishifune C, Maekawa Y, Koyanagi A, Yagita H, et al. Dendritic cell-mediated NK cell activation is controlled by Jagged2-Notch interaction. Proc Natl Acad Sci U S A. 2008;105(19):7010–5. doi: 10.1073/pnas.0709919105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Haraguchi K, Suzuki T, Koyama N, Kumano K, Nakahara F, Matsumoto A, et al. Notch activation induces the generation of functional NK cells from human cord blood CD34-positive cells devoid of IL-15. J Immunol. 2009;182(10):6168–78. doi: 10.4049/jimmunol.0803036. [DOI] [PubMed] [Google Scholar]
  • 109.Ohishi K, Varnum-Finney B, Serda RE, Anasetti C, Bernstein ID. The Notch ligand, Delta-1, inhibits the differentiation of monocytes into macrophages but permits their differentiation into dendritic cells. Blood. 2001;98(5):1402–7. doi: 10.1182/blood.v98.5.1402. [DOI] [PubMed] [Google Scholar]
  • 110.Cheng P, Gabrilovich D. Notch signaling in differentiation and function of dendritic cells. Immunol Res. 2008;41(1):1–14. doi: 10.1007/s12026-007-8011-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Han H, Tanigaki K, Yamamoto N, Kuroda K, Yoshimoto M, Nakahata T, et al. Inducible gene knockout of transcription factor recombination signal binding protein-J reveals its essential role in T versus B lineage decision. Int Immunol. 2002;14(6):637–45. doi: 10.1093/intimm/dxf030. [DOI] [PubMed] [Google Scholar]
  • 112.Izon DJ, Punt JA, Pear WS. Deciphering the role of Notch signaling in lymphopoiesis. Curr Opin Immunol. 2002;14(2):192–9. doi: 10.1016/s0952-7915(02)00321-7. [DOI] [PubMed] [Google Scholar]
  • 113.Radtke F, Wilson A, Stark G, Bauer M, van Meerwijk J, MacDonald HR, et al. Deficient T cell fate specification in mice with an induced inactivation of Notch1. Immunity. 1999;10(5):547–58. doi: 10.1016/s1074-7613(00)80054-0. [DOI] [PubMed] [Google Scholar]
  • 114.Koch U, Lacombe TA, Holland D, Bowman JL, Cohen BL, Egan SE, et al. Subversion of the T/B lineage decision in the thymus by lunatic fringe-mediated inhibition of Notch-1. Immunity. 2001;15(2):225–36. doi: 10.1016/s1074-7613(01)00189-3. [DOI] [PubMed] [Google Scholar]
  • 115.Doerfler P, Shearman MS, Perlmutter RM. Presenilin-dependent gamma-secretase activity modulates thymocyte development. Proc Natl Acad Sci U S A. 2001;98(16):9312–7. doi: 10.1073/pnas.161102498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Hadland BK, Manley NR, Su D, Longmore GD, Moore CL, Wolfe MS, et al. Gamma -secretase inhibitors repress thymocyte development. Proc Natl Acad Sci U S A. 2001;98(13):7487–91. doi: 10.1073/pnas.131202798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.De Smedt M, Hoebeke I, Reynvoet K, Leclercq G, Plum J. Different thresholds of Notch signaling bias human precursor cells toward B-, NK-, monocytic/dendritic-, or T-cell lineage in thymus microenvironment. Blood. 2005;106(10):3498–506. doi: 10.1182/blood-2005-02-0496. [DOI] [PubMed] [Google Scholar]
  • 118.Hozumi K, Mailhos C, Negishi N, Hirano K, Yahata T, Ando K, et al. Delta-like 4 is indispensable in thymic environment specific for T cell development. J Exp Med. 2008;205(11):2507–13. doi: 10.1084/jem.20080134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Koch U, Fiorini E, Benedito R, Besseyrias V, Schuster-Gossler K, Pierres M, et al. Delta-like 4 is the essential, nonredundant ligand for Notch1 during thymic T cell lineage commitment. J Exp Med. 2008;205(11):2515–23. doi: 10.1084/jem.20080829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Maeda T, Merghoub T, Hobbs RM, Dong L, Maeda M, Zakrzewski J, et al. Regulation of B versus T lymphoid lineage fate decision by the proto-oncogene LRF. Science. 2007;316(5826):860–6. doi: 10.1126/science.1140881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Bell JJ, Bhandoola A. The earliest thymic progenitors for T cells possess myeloid lineage potential. Nature. 2008;452(7188):764–7. doi: 10.1038/nature06840. [DOI] [PubMed] [Google Scholar]
  • 122.Feyerabend TB, Terszowski G, Tietz A, Blum C, Luche H, Gossler A, et al. Deletion of Notch1 converts pro-T cells to dendritic cells and promotes thymic B cells by cell-extrinsic and cell-intrinsic mechanisms. Immunity. 2009;30(1):67–79. doi: 10.1016/j.immuni.2008.10.016. [DOI] [PubMed] [Google Scholar]
  • 123.Kumano K, Chiba S, Kunisato A, Sata M, Saito T, Nakagami-Yamaguchi E, et al. Notch1 but not Notch2 is essential for generating hematopoietic stem cells from endothelial cells. Immunity. 2003;18(5):699–711. doi: 10.1016/s1074-7613(03)00117-1. [DOI] [PubMed] [Google Scholar]
  • 124.Robert-Moreno A, Guiu J, Ruiz-Herguido C, Lopez ME, Ingles-Esteve J, Riera L, et al. Impaired embryonic haematopoiesis yet normal arterial development in the absence of the Notch ligand Jagged1. Embo J. 2008;27(13):1886–95. doi: 10.1038/emboj.2008.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Hadland BK, Huppert SS, Kanungo J, Xue Y, Jiang R, Gridley T, et al. A requirement for Notch1 distinguishes 2 phases of definitive hematopoiesis during development. Blood. 2004;104(10):3097–105. doi: 10.1182/blood-2004-03-1224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, Knight MC, et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425(6960):841–6. doi: 10.1038/nature02040. [DOI] [PubMed] [Google Scholar]
  • 127.Kunisato A, Chiba S, Nakagami-Yamaguchi E, Kumano K, Saito T, Masuda S, et al. HES-1 preserves purified hematopoietic stem cells ex vivo and accumulates side population cells in vivo. Blood. 2003;101(5):1777–83. doi: 10.1182/blood-2002-07-2051. [DOI] [PubMed] [Google Scholar]
  • 128.Stier S, Cheng T, Dombkowski D, Carlesso N, Scadden DT. Notch1 activation increases hematopoietic stem cell self-renewal in vivo and favors lymphoid over myeloid lineage outcome. Blood. 2002;99(7):2369–78. doi: 10.1182/blood.v99.7.2369. [DOI] [PubMed] [Google Scholar]
  • 129.Maillard I, Koch U, Dumortier A, Shestova O, Xu L, Sai H, et al. Canonical notch signaling is dispensable for the maintenance of adult hematopoietic stem cells. Cell Stem Cell. 2008;2(4):356–66. doi: 10.1016/j.stem.2008.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Blanpain C, Fuchs E. Epidermal homeostasis: a balancing act of stem cells in the skin. Nat Rev Mol Cell Biol. 2009;10(3):207–17. doi: 10.1038/nrm2636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Fuchs E, Raghavan S. Getting under the skin of epidermal morphogenesis. Nat Rev Genet. 2002;3(3):199–209. doi: 10.1038/nrg758. [DOI] [PubMed] [Google Scholar]
  • 132.Ambler CA, Maatta A. Epidermal stem cells: location, potential and contribution to cancer. J Pathol. 2009;217(2):206–16. doi: 10.1002/path.2468. [DOI] [PubMed] [Google Scholar]
  • 133.Okuyama R, Tagami H, Aiba S. Notch signaling: its role in epidermal homeostasis and in the pathogenesis of skin diseases. J Dermatol Sci. 2008;49(3):187–94. doi: 10.1016/j.jdermsci.2007.05.017. [DOI] [PubMed] [Google Scholar]
  • 134.Watt FM, Estrach S, Ambler CA. Epidermal Notch signalling: differentiation, cancer and adhesion. Curr Opin Cell Biol. 2008;20(2):171–9. doi: 10.1016/j.ceb.2008.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Blanpain C, Lowry WE, Pasolli HA, Fuchs E. Canonical notch signaling functions as a commitment switch in the epidermal lineage. Genes Dev. 2006;20(21):3022–35. doi: 10.1101/gad.1477606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Rangarajan A, Talora C, Okuyama R, Nicolas M, Mammucari C, Oh H, et al. Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation. Embo J. 2001;20(13):3427–36. doi: 10.1093/emboj/20.13.3427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Demehri S, Turkoz A, Kopan R. Epidermal Notch1 loss promotes skin tumorigenesis by impacting the stromal microenvironment. Cancer Cell. 2009;16(1):55–66. doi: 10.1016/j.ccr.2009.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Pan Y, Lin MH, Tian X, Cheng HT, Gridley T, Shen J, et al. gamma-secretase functions through Notch signaling to maintain skin appendages but is not required for their patterning or initial morphogenesis. Dev Cell. 2004;7(5):731–43. doi: 10.1016/j.devcel.2004.09.014. [DOI] [PubMed] [Google Scholar]
  • 139.Nicolas M, Wolfer A, Raj K, Kummer JA, Mill P, van Noort M, et al. Notch1 functions as a tumor suppressor in mouse skin. Nat Genet. 2003;33(3):416–21. doi: 10.1038/ng1099. [DOI] [PubMed] [Google Scholar]
  • 140.Dotto GP. Notch tumor suppressor function. Oncogene. 2008;27(38):5115–23. doi: 10.1038/onc.2008.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Li T, Wen H, Brayton C, Laird FM, Ma G, Peng S, et al. Moderate reduction of gamma-secretase attenuates amyloid burden and limits mechanism-based liabilities. J Neurosci. 2007;27(40):10849–59. doi: 10.1523/JNEUROSCI.2152-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Tournoy J, Bossuyt X, Snellinx A, Regent M, Garmyn M, Serneels L, et al. Partial loss of presenilins causes seborrheic keratosis and autoimmune disease in mice. Hum Mol Genet. 2004;13(13):1321–31. doi: 10.1093/hmg/ddh151. [DOI] [PubMed] [Google Scholar]
  • 143.Demehri S, Liu Z, Lee J, Lin MH, Crosby SD, Roberts CJ, et al. Notch-deficient skin induces a lethal systemic B-lymphoproliferative disorder by secreting TSLP, a sentinel for epidermal integrity. PLoS Biol. 2008;6(5):e123. doi: 10.1371/journal.pbio.0060123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Dumortier A, Durham AD, Di Piazza M, Vauclair S, Koch U, Ferrand G, et al. Atopic dermatitis-like disease and associated lethal myeloproliferative disorder arise from loss of notch signaling in the murine skin. PLoS One. 5(2):e9258. doi: 10.1371/journal.pone.0009258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Qyang Y, Chambers SM, Wang P, Xia X, Chen X, Goodell MA, et al. Myeloproliferative disease in mice with reduced presenilin gene dosage: effect of gamma-secretase blockage. Biochemistry. 2004;43(18):5352–9. doi: 10.1021/bi049826u. [DOI] [PubMed] [Google Scholar]
  • 146.Wang B, Yang W, Wen W, Sun J, Su B, Liu B, et al. {gamma}-Secretase Gene Mutations in Familial Acne Inversa. Science. 2010 doi: 10.1126/science.1196284. [DOI] [PubMed] [Google Scholar]
  • 147.Lledo PM, Alonso M, Grubb MS. Adult neurogenesis and functional plasticity in neuronal circuits. Nat Rev Neurosci. 2006;7(3):179–93. doi: 10.1038/nrn1867. [DOI] [PubMed] [Google Scholar]
  • 148.Suh H, Deng W, Gage FH. Signaling in adult neurogenesis. Annu Rev Cell Dev Biol. 2009;25(253-75) doi: 10.1146/annurev.cellbio.042308.113256. [DOI] [PubMed] [Google Scholar]
  • 149.Louvi A, Artavanis-Tsakonas S. Notch signalling in vertebrate neural development. Nat Rev Neurosci. 2006;7(2):93–102. doi: 10.1038/nrn1847. [DOI] [PubMed] [Google Scholar]
  • 150.Cau E, Blader P. Notch activity in the nervous system: to switch or not switch? Neural Dev. 2009;4(36) doi: 10.1186/1749-8104-4-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Johnson MA, Ables JL, Eisch AJ. Cell-intrinsic signals that regulate adult neurogenesis in vivo: insights from inducible approaches. BMB Rep. 2009;42(5):245–59. doi: 10.5483/bmbrep.2009.42.5.245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Breunig JJ, Silbereis J, Vaccarino FM, Sestan N, Rakic P. Notch regulates cell fate and dendrite morphology of newborn neurons in the postnatal dentate gyrus. Proc Natl Acad Sci U S A. 2007;104(51):20558–63. doi: 10.1073/pnas.0710156104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Ehm O, Goritz C, Covic M, Schaffner I, Schwarz TJ, Karaca E, et al. RBPJkappa-dependent signaling is essential for long-term maintenance of neural stem cells in the adult hippocampus. J Neurosci. 2010;30(41):13794–807. doi: 10.1523/JNEUROSCI.1567-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Imayoshi I, Sakamoto M, Yamaguchi M, Mori K, Kageyama R. Essential roles of Notch signaling in maintenance of neural stem cells in developing and adult brains. J Neurosci. 30(9):3489–98. doi: 10.1523/JNEUROSCI.4987-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Carlen M, Meletis K, Goritz C, Darsalia V, Evergren E, Tanigaki K, et al. Forebrain ependymal cells are Notch-dependent and generate neuroblasts and astrocytes after stroke. Nat Neurosci. 2009;12(3):259–67. doi: 10.1038/nn.2268. [DOI] [PubMed] [Google Scholar]
  • 156.Lathia JD, Mattson MP, Cheng A. Notch: from neural development to neurological disorders. J Neurochem. 2008;107(6):1471–81. doi: 10.1111/j.1471-4159.2008.05715.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Berezovska O, Frosch M, McLean P, Knowles R, Koo E, Kang D, et al. The Alzheimer-related gene presenilin 1 facilitates notch 1 in primary mammalian neurons. Brain Res Mol Brain Res. 1999;69(2):273–80. doi: 10.1016/s0169-328x(99)00119-9. [DOI] [PubMed] [Google Scholar]
  • 158.Sestan N, Artavanis-Tsakonas S, Rakic P. Contact-dependent inhibition of cortical neurite growth mediated by notch signaling. Science. 1999;286(5440):741–6. doi: 10.1126/science.286.5440.741. [DOI] [PubMed] [Google Scholar]
  • 159.Wang Y, Chan SL, Miele L, Yao PJ, Mackes J, Ingram DK, et al. Involvement of Notch signaling in hippocampal synaptic plasticity. Proc Natl Acad Sci U S A. 2004;101(25):9458–62. doi: 10.1073/pnas.0308126101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Ge X, Hannan F, Xie Z, Feng C, Tully T, Zhou H, et al. Notch signaling in Drosophila long-term memory formation. Proc Natl Acad Sci U S A. 2004;101(27):10172–6. doi: 10.1073/pnas.0403497101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Presente A, Boyles RS, Serway CN, de Belle JS, Andres AJ. Notch is required for long-term memory in Drosophila. Proc Natl Acad Sci U S A. 2004;101(6):1764–8. doi: 10.1073/pnas.0308259100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Saura CA, Choi SY, Beglopoulos V, Malkani S, Zhang D, Shankaranarayana Rao BS, et al. Loss of presenilin function causes impairments of memory and synaptic plasticity followed by age-dependent neurodegeneration. Neuron. 2004;42(1):23–36. doi: 10.1016/s0896-6273(04)00182-5. [DOI] [PubMed] [Google Scholar]
  • 163.Tabuchi K, Chen G, Sudhof TC, Shen J. Conditional forebrain inactivation of nicastrin causes progressive memory impairment and age-related neurodegeneration. J Neurosci. 2009;29(22):7290–301. doi: 10.1523/JNEUROSCI.1320-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Yu H, Saura CA, Choi SY, Sun LD, Yang X, Handler M, et al. APP processing and synaptic plasticity in presenilin-1 conditional knockout mice. Neuron. 2001;31(5):713–26. doi: 10.1016/s0896-6273(01)00417-2. [DOI] [PubMed] [Google Scholar]
  • 165.Costa RM, Honjo T, Silva AJ. Learning and memory deficits in Notch mutant mice. Curr Biol. 2003;13(15):1348–54. doi: 10.1016/s0960-9822(03)00492-5. [DOI] [PubMed] [Google Scholar]
  • 166.Lazarov O, Marr RA. Neurogenesis and Alzheimer's disease: At the crossroads. Exp Neurol. 2009 doi: 10.1016/j.expneurol.2009.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Marlatt MW, Lucassen PJ. Neurogenesis and Alzheimer's disease: Biology and pathophysiology in mice and men. Curr Alzheimer Res. 7(2):113–25. doi: 10.2174/156720510790691362. [DOI] [PubMed] [Google Scholar]
  • 168.Taupin P. Adult neurogenesis, neural stem cells and Alzheimer's disease: developments, limitations, problems and promises. Curr Alzheimer Res. 2009;6(6):461–70. doi: 10.2174/156720509790147151. [DOI] [PubMed] [Google Scholar]
  • 169.Woo HN, Park JS, Gwon AR, Arumugam TV, Jo DG. Alzheimer's disease and Notch signaling. Biochem Biophys Res Commun. 2009;390(4):1093–7. doi: 10.1016/j.bbrc.2009.10.093. [DOI] [PubMed] [Google Scholar]
  • 170.Zammit PS. All muscle satellite cells are equal, but are some more equal than others? J Cell Sci. 2008;121(Pt 18):2975–82. doi: 10.1242/jcs.019661. [DOI] [PubMed] [Google Scholar]
  • 171.Schuster-Gossler K, Cordes R, Gossler A. Premature myogenic differentiation and depletion of progenitor cells cause severe muscle hypotrophy in Delta1 mutants. Proc Natl Acad Sci U S A. 2007;104(2):537–42. doi: 10.1073/pnas.0608281104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Carlson ME, Conboy IM. Regulating the Notch pathway in embryonic, adult and old stem cells. Curr Opin Pharmacol. 2007;7(3):303–9. doi: 10.1016/j.coph.2007.02.004. [DOI] [PubMed] [Google Scholar]
  • 173.Conboy IM, Rando TA. Aging, stem cells and tissue regeneration: lessons from muscle. Cell Cycle. 2005;4(3):407–10. doi: 10.4161/cc.4.3.1518. [DOI] [PubMed] [Google Scholar]
  • 174.Luo D, Renault VM, Rando TA. The regulation of Notch signaling in muscle stem cell activation and postnatal myogenesis. Semin Cell Dev Biol. 2005;16(4-5):612–22. doi: 10.1016/j.semcdb.2005.07.002. [DOI] [PubMed] [Google Scholar]
  • 175.Conboy IM, Rando TA. The regulation of Notch signaling controls satellite cell activation and cell fate determination in postnatal myogenesis. Dev Cell. 2002;3(3):397–409. doi: 10.1016/s1534-5807(02)00254-x. [DOI] [PubMed] [Google Scholar]
  • 176.Kuang S, Kuroda K, Le Grand F, Rudnicki MA. Asymmetric self-renewal and commitment of satellite stem cells in muscle. Cell. 2007;129(5):999–1010. doi: 10.1016/j.cell.2007.03.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Brack AS, Conboy IM, Conboy MJ, Shen J, Rando TA. A temporal switch from notch to Wnt signaling in muscle stem cells is necessary for normal adult myogenesis. Cell Stem Cell. 2008;2(1):50–9. doi: 10.1016/j.stem.2007.10.006. [DOI] [PubMed] [Google Scholar]
  • 178.Conboy IM, Conboy MJ, Smythe GM, Rando TA. Notch-mediated restoration of regenerative potential to aged muscle. Science. 2003;302(5650):1575–7. doi: 10.1126/science.1087573. [DOI] [PubMed] [Google Scholar]
  • 179.Conboy IM, Conboy MJ, Wagers AJ, Girma ER, Weissman IL, Rando TA. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature. 2005;433(7027):760–4. doi: 10.1038/nature03260. [DOI] [PubMed] [Google Scholar]
  • 180.Brown D, Hikim AP, Kovacheva EL, Sinha-Hikim I. Mouse model of testosterone-induced muscle fiber hypertrophy: involvement of p38 mitogen-activated protein kinase-mediated Notch signaling. J Endocrinol. 2009;201(1):129–39. doi: 10.1677/JOE-08-0476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Carlson ME, Conboy MJ, Hsu M, Barchas L, Jeong J, Agrawal A, et al. Relative roles of TGF-beta1 and Wnt in the systemic regulation and aging of satellite cell responses. Aging Cell. 2009;8(6):676–89. doi: 10.1111/j.1474-9726.2009.00517.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Gridley T. Notch signaling and inherited disease syndromes. Hum Mol Genet. 2003;12(R9-13) doi: 10.1093/hmg/ddg052. Spec No 1. [DOI] [PubMed] [Google Scholar]
  • 183.Ellisen LW, Bird J, West DC, Soreng AL, Reynolds TC, Smith SD, et al. TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms. Cell. 1991;66(4):649–61. doi: 10.1016/0092-8674(91)90111-b. [DOI] [PubMed] [Google Scholar]
  • 184.Real PJ, Ferrando AA. NOTCH inhibition and glucocorticoid therapy in T-cell acute lymphoblastic leukemia. Leukemia. 2009;23(8):1374–7. doi: 10.1038/leu.2009.75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Aifantis I, Raetz E, Buonamici S. Molecular pathogenesis of T-cell leukaemia and lymphoma. Nat Rev Immunol. 2008;8(5):380–90. doi: 10.1038/nri2304. [DOI] [PubMed] [Google Scholar]
  • 186.Ferrando AA, Neuberg DS, Staunton J, Loh ML, Huard C, Raimondi SC, et al. Gene expression signatures define novel oncogenic pathways in T cell acute lymphoblastic leukemia. Cancer Cell. 2002;1(1):75–87. doi: 10.1016/s1535-6108(02)00018-1. [DOI] [PubMed] [Google Scholar]
  • 187.Pear WS, Aster JC. T cell acute lymphoblastic leukemia/lymphoma: a human cancer commonly associated with aberrant NOTCH1 signaling. Curr Opin Hematol. 2004;11(6):426–33. doi: 10.1097/01.moh.0000143965.90813.70. [DOI] [PubMed] [Google Scholar]
  • 188.Gridley T. Kick it up a Notch: NOTCH1 activation in T-ALL. Cancer Cell. 2004;6(5):431–2. doi: 10.1016/j.ccr.2004.10.019. [DOI] [PubMed] [Google Scholar]
  • 189.Chiang MY, Xu ML, Histen G, Shestova O, Roy M, Nam Y, et al. Identification of a conserved negative regulatory sequence that influences the leukemogenic activity of NOTCH1. Mol Cell Biol. 2006;26(16):6261–71. doi: 10.1128/MCB.02478-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.O'Neil J, Grim J, Strack P, Rao S, Tibbitts D, Winter C, et al. FBW7 mutations in leukemic cells mediate NOTCH pathway activation and resistance to gamma-secretase inhibitors. J Exp Med. 2007;204(8):1813–24. doi: 10.1084/jem.20070876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Sulis ML, Williams O, Palomero T, Tosello V, Pallikuppam S, Real PJ, et al. NOTCH1 extracellular juxtamembrane expansion mutations in T-ALL. Blood. 2008;112(3):733–40. doi: 10.1182/blood-2007-12-130096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Palomero T, Lim WK, Odom DT, Sulis ML, Real PJ, Margolin A, et al. NOTCH1 directly regulates c-MYC and activates a feed-forward-loop transcriptional network promoting leukemic cell growth. Proc Natl Acad Sci U S A. 2006;103(48):18261–6. doi: 10.1073/pnas.0606108103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Sharma VM, Calvo JA, Draheim KM, Cunningham LA, Hermance N, Beverly L, et al. Notch1 contributes to mouse T-cell leukemia by directly inducing the expression of c-myc. Mol Cell Biol. 2006;26(21):8022–31. doi: 10.1128/MCB.01091-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Weng AP, Lau A. Notch signaling in T-cell acute lymphoblastic leukemia. Future Oncol. 2005;1(4):511–9. doi: 10.2217/14796694.1.4.511. [DOI] [PubMed] [Google Scholar]
  • 195.Vilimas T, Mascarenhas J, Palomero T, Mandal M, Buonamici S, Meng F, et al. Targeting the NF-kappaB signaling pathway in Notch1-induced T-cell leukemia. Nat Med. 2007;13(1):70–7. doi: 10.1038/nm1524. [DOI] [PubMed] [Google Scholar]
  • 196.Palomero T, Dominguez M, Ferrando AA. The role of the PTEN/AKT Pathway in NOTCH1-induced leukemia. Cell Cycle. 2008;7(8):965–70. doi: 10.4161/cc.7.8.5753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Medyouf H, Ghysdael J. The calcineurin/NFAT signaling pathway: a novel therapeutic target in leukemia and solid tumors. Cell Cycle. 2008;7(3):297–303. doi: 10.4161/cc.7.3.5357. [DOI] [PubMed] [Google Scholar]
  • 198.Palomero T, Sulis ML, Cortina M, Real PJ, Barnes K, Ciofani M, et al. Mutational loss of PTEN induces resistance to NOTCH1 inhibition in T-cell leukemia. Nat Med. 2007;13(10):1203–10. doi: 10.1038/nm1636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Tournier-Lasserve E, Joutel A, Melki J, Weissenbach J, Lathrop GM, Chabriat H, et al. Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy maps to chromosome 19q12. Nat Genet. 1993;3(3):256–9. doi: 10.1038/ng0393-256. [DOI] [PubMed] [Google Scholar]
  • 200.Chabriat H, Joutel A, Dichgans M, Tournier-Lasserve E, Bousser MG. Cadasil. Lancet Neurol. 2009;8(7):643–53. doi: 10.1016/S1474-4422(09)70127-9. [DOI] [PubMed] [Google Scholar]
  • 201.Joutel A, Corpechot C, Ducros A, Vahedi K, Chabriat H, Mouton P, et al. Notch3 mutations in CADASIL, a hereditary adult-onset condition causing stroke and dementia. Nature. 1996;383(6602):707–10. doi: 10.1038/383707a0. [DOI] [PubMed] [Google Scholar]
  • 202.Domenga V, Fardoux P, Lacombe P, Monet M, Maciazek J, Krebs LT, et al. Notch3 is required for arterial identity and maturation of vascular smooth muscle cells. Genes Dev. 2004;18(22):2730–5. doi: 10.1101/gad.308904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Joutel A, Andreux F, Gaulis S, Domenga V, Cecillon M, Battail N, et al. The ectodomain of the Notch3 receptor accumulates within the cerebrovasculature of CADASIL patients. J Clin Invest. 2000;105(5):597–605. doi: 10.1172/JCI8047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Tikka S, Mykkanen K, Ruchoux MM, Bergholm R, Junna M, Poyhonen M, et al. Congruence between NOTCH3 mutations and GOM in 131 CADASIL patients. Brain. 2009;132(Pt 4):933–9. doi: 10.1093/brain/awn364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Fryxell KJ, Soderlund M, Jordan TV. An animal model for the molecular genetics of CADASIL. (Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy). Stroke. 2001;32(1):6–11. doi: 10.1161/01.str.32.1.6. [DOI] [PubMed] [Google Scholar]
  • 206.Spinner NB. CADASIL: Notch signaling defect or protein accumulation problem? J Clin Invest. 2000;105(5):561–2. doi: 10.1172/JCI9511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Rampal R, Luther KB, Haltiwanger RS. Notch signaling in normal and disease States: possible therapies related to glycosylation. Curr Mol Med. 2007;7(4):427–45. doi: 10.2174/156652407780831593. [DOI] [PubMed] [Google Scholar]
  • 208.Karlstrom H, Beatus P, Dannaeus K, Chapman G, Lendahl U, Lundkvist J. A CADASIL-mutated Notch 3 receptor exhibits impaired intracellular trafficking and maturation but normal ligand-induced signaling. Proc Natl Acad Sci U S A. 2002;99(26):17119–24. doi: 10.1073/pnas.252624099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Joutel A, Monet M, Domenga V, Riant F, Tournier-Lasserve E. Pathogenic mutations associated with cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy differently affect Jagged1 binding and Notch3 activity via the RBP/JK signaling Pathway. Am J Hum Genet. 2004;74(2):338–47. doi: 10.1086/381506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Arboleda-Velasquez JF, Lopera F, Lopez E, Frosch MP, Sepulveda-Falla D, Gutierrez JE, et al. C455R notch3 mutation in a Colombian CADASIL kindred with early onset of stroke. Neurology. 2002;59(2):277–9. doi: 10.1212/wnl.59.2.277. [DOI] [PubMed] [Google Scholar]
  • 211.Belin de Chantemele EJ, Retailleau K, Pinaud F, Vessieres E, Bocquet A, Guihot AL, et al. Notch3 is a major regulator of vascular tone in cerebral and tail resistance arteries. Arterioscler Thromb Vasc Biol. 2008;28(12):2216–24. doi: 10.1161/ATVBAHA.108.171751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Arboleda-Velasquez JF, Zhou Z, Shin HK, Louvi A, Kim HH, Savitz SI, et al. Linking Notch signaling to ischemic stroke. Proc Natl Acad Sci U S A. 2008;105(12):4856–61. doi: 10.1073/pnas.0709867105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Monet M, Domenga V, Lemaire B, Souilhol C, Langa F, Babinet C, et al. The archetypal R90C CADASIL-NOTCH3 mutation retains NOTCH3 function in vivo. Hum Mol Genet. 2007;16(8):982–92. doi: 10.1093/hmg/ddm042. [DOI] [PubMed] [Google Scholar]
  • 214.Opherk C, Duering M, Peters N, Karpinska A, Rosner S, Schneider E, et al. CADASIL mutations enhance spontaneous multimerization of NOTCH3. Hum Mol Genet. 2009;18(15):2761–7. doi: 10.1093/hmg/ddp211. [DOI] [PubMed] [Google Scholar]
  • 215.Takahashi K, Adachi K, Yoshizaki K, Kunimoto S, Kalaria RN, Watanabe A. Mutations in NOTCH3 cause the formation and retention of aggregates in the endoplasmic reticulum, leading to impaired cell proliferation. Hum Mol Genet. 19(1):79–89. doi: 10.1093/hmg/ddp468. [DOI] [PubMed] [Google Scholar]
  • 216.Joutel A, Monet-Lepretre M, Gosele C, Baron-Menguy C, Hammes A, Schmidt S, et al. Cerebrovascular dysfunction and microcirculation rarefaction precede white matter lesions in a mouse genetic model of cerebral ischemic small vessel disease. J Clin Invest. 120(2):433–45. doi: 10.1172/JCI39733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Hohlfeld R. Myelin failure in multiple sclerosis: breaking the spell of Notch. Nat Med. 2002;8(10):1075–6. doi: 10.1038/nm1002-1075. [DOI] [PubMed] [Google Scholar]
  • 218.Brosnan CF, John GR. Revisiting Notch in remyelination of multiple sclerosis lesions. J Clin Invest. 2009;119(1):10–3. doi: 10.1172/JCI37786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Jurynczyk M, Selmaj K. Notch: a new player in MS mechanisms. J Neuroimmunol. 2010;218(1-2):3–11. doi: 10.1016/j.jneuroim.2009.08.010. [DOI] [PubMed] [Google Scholar]
  • 220.Franklin RJ. Why does remyelination fail in multiple sclerosis? Nat Rev Neurosci. 2002;3(9):705–14. doi: 10.1038/nrn917. [DOI] [PubMed] [Google Scholar]
  • 221.Wang S, Sdrulla AD, diSibio G, Bush G, Nofziger D, Hicks C, et al. Notch receptor activation inhibits oligodendrocyte differentiation. Neuron. 1998;21(1):63–75. doi: 10.1016/s0896-6273(00)80515-2. [DOI] [PubMed] [Google Scholar]
  • 222.Genoud S, Lappe-Siefke C, Goebbels S, Radtke F, Aguet M, Scherer SS, et al. Notch1 control of oligodendrocyte differentiation in the spinal cord. J Cell Biol. 2002;158(4):709–18. doi: 10.1083/jcb.200202002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Blaschuk KL, ffrench-Constant C. Developmental neurobiology: notch is tops in the developing brain. Curr Biol. 1998;8(10):R334–7. doi: 10.1016/s0960-9822(98)70215-5. [DOI] [PubMed] [Google Scholar]
  • 224.John GR, Shankar SL, Shafit-Zagardo B, Massimi A, Lee SC, Raine CS, et al. Multiple sclerosis: re-expression of a developmental pathway that restricts oligodendrocyte maturation. Nat Med. 2002;8(10):1115–21. doi: 10.1038/nm781. [DOI] [PubMed] [Google Scholar]
  • 225.Seifert T, Bauer J, Weissert R, Fazekas F, Storch MK. Notch1 and its ligand Jagged1 are present in remyelination in a T-cell- and antibody-mediated model of inflammatory demyelination. Acta Neuropathol. 2007;113(2):195–203. doi: 10.1007/s00401-006-0170-9. [DOI] [PubMed] [Google Scholar]
  • 226.Stidworthy MF, Genoud S, Li WW, Leone DP, Mantei N, Suter U, et al. Notch1 and Jagged1 are expressed after CNS demyelination, but are not a major rate-determining factor during remyelination. Brain. 2004;127(Pt 9):1928–41. doi: 10.1093/brain/awh217. [DOI] [PubMed] [Google Scholar]
  • 227.Zhang Y, Argaw AT, Gurfein BT, Zameer A, Snyder BJ, Ge C, et al. Notch1 signaling plays a role in regulating precursor differentiation during CNS remyelination. Proc Natl Acad Sci U S A. 2009;106(45):19162–7. doi: 10.1073/pnas.0902834106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Hu QD, Ang BT, Karsak M, Hu WP, Cui XY, Duka T, et al. F3/contactin acts as a functional ligand for Notch during oligodendrocyte maturation. Cell. 2003;115(2):163–75. doi: 10.1016/s0092-8674(03)00810-9. [DOI] [PubMed] [Google Scholar]
  • 229.Nakahara J, Kanekura K, Nawa M, Aiso S, Suzuki N. Abnormal expression of TIP30 and arrested nucleocytoplasmic transport within oligodendrocyte precursor cells in multiple sclerosis. J Clin Invest. 2009;119(1):169–81. doi: 10.1172/JCI35440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Gorlich D, Vogel F, Mills AD, Hartmann E, Laskey RA. Distinct functions for the two importin subunits in nuclear protein import. Nature. 1995;377(6546):246–8. doi: 10.1038/377246a0. [DOI] [PubMed] [Google Scholar]
  • 231.Fancy SP, Kotter MR, Harrington EP, Huang JK, Zhao C, Rowitch DH, et al. Overcoming remyelination failure in multiple sclerosis and other myelin disorders. Exp Neurol. doi: 10.1016/j.expneurol.2009.12.020. [DOI] [PubMed] [Google Scholar]
  • 232.Jurynczyk M, Jurewicz A, Bielecki B, Raine CS, Selmaj K. Inhibition of Notch signaling enhances tissue repair in an animal model of multiple sclerosis. J Neuroimmunol. 2005;170(1-2):3–10. doi: 10.1016/j.jneuroim.2005.10.013. [DOI] [PubMed] [Google Scholar]
  • 233.Jurynczyk M, Jurewicz A, Raine CS, Selmaj K. Notch3 inhibition in myelin-reactive T cells down-regulates protein kinase C theta and attenuates experimental autoimmune encephalomyelitis. J Immunol. 2008;180(4):2634–40. doi: 10.4049/jimmunol.180.4.2634. [DOI] [PubMed] [Google Scholar]
  • 234.Minter LM, Turley DM, Das P, Shin HM, Joshi I, Lawlor RG, et al. Inhibitors of gamma-secretase block in vivo and in vitro T helper type 1 polarization by preventing Notch upregulation of Tbx21. Nat Immunol. 2005;6(7):680–8. [PubMed] [Google Scholar]

RESOURCES