Notch is a well-conserved signaling pathway and its function in cell fate determination is crucial in embryonic development and in the maintenance of tissue homeostasis during adult life. Notch activation depends on cell-cell interactions that are essential for the generation of cell diversity from initially equivalent cell populations. In the adult hematopoiesis, Notch is undoubtedly a very efficient promoter of T-cell differentiation, and this has masked for a long time the effects of Notch on other blood lineages, which are gradually being identified. However, the adult hematopoietic stem cell (HSC) remains mostly refractory to Notch intervention in experimental systems. In contrast, Notch is essential for the generation of the HSCs, which takes place during embryonic development. This review summarizes the knowledge accumulated in recent years regarding the role of the Notch pathway in the different stages of HSC ontology from embryonic life to fetal and adult bone marrow stem cells. In addition, we briefly examine other systems where Notch regulates specific stem cell capacities, in an attempt to understand how Notch functions in stem cell biology.

Stem cell biology is dependent on a few signaling pathways that crosstalk each other to generate a network of biochemical interactions and are ultimately translated into precise instructions at the nuclear level. The Notch pathway has been evolutionary preserved, not only biochemically but also functionally, being involved in establishing cell diversification from equipotent adjacent cells using a mechanism that requires cell-cell interaction. Essentially, Notch-mediated communication depends on the differential expression of specific ligands (Jagged or Delta) and receptors (Notch1-4) in adjacent cells. Basically, Notch signaling is transduced from the sending cell (the one that expresses higher levels of the ligand) to the nucleus of the receiving cell (expressing higher levels of Notch receptors), thus impinging on its biochemical network.1  Cell-cell interactions are essential players in the regulation of stem cell and tissue homeostasis of the adult but also in developing organisms.

In general, the tag “stem” refers to cells with unlimited capacity for self-renewal and with the ability to generate all the different lineages of a specific system. However, when studying stem cells, one needs to distinguish 2 main categories of stem cells: (1) embryonic stem cells, which are pluripotent and retain the capacity to generate all the cell lineages of the adult organism; and (2) somatic stem cells, which are also generated in the developing embryo, maintain the self renewal capacity, but show a reduced pluripotency because they can only generate a limited number of cell types. The latter include the ones involved in tissue formation and regeneration both in the embryo and the adult.2 

The hematopoietic system has served for decades as a pioneer model for studying and deciphering the behavior of somatic stem cells and the mechanisms that regulate cell differentiation; and indeed, the knowledge generated on hematopoietic stem cell (HSC) biology has guided the understanding of other types of somatic stem cells. From these studies, the Notch pathway has emerged as a principal player on stem cell regulation and differentiation, even though many questions on how or whether Notch functions in HSCs remain unanswered. Interestingly, studies in the adult HSCs unexpectedly revealed that the maintenance of this type of stem cells was independent of Notch. However, further understanding of the ontogeny of HSCs has now provided new insights about the predominant role of Notch in the generation of HSCs in the embryo. The aim of this review is to gain a better understanding of the Notch pathway and integrate the results obtained in the last decades by analyzing HSCs from the adult bone marrow and the mouse embryo.

Notch is a well-conserved signaling pathway that was first identified in Drosophila mutants. Lack of the X-linked Notch locus in the fly resulted in embryonic death, yet heterozygous females showed the serrate/notched wing margin first observed by T. H. Morgan's group,3  a phenotype that named the whole pathway. The Notch gene was initially cloned in Drosophila4,5  and soon after in Caenorhabditis elegans,6,7  Xenopus,8  mouse,9  and human.10  In general, Notch works as a determinant factor during binary cell decisions from adjacent cells, a function that was elegantly demonstrated in C elegans11  and Drosophila.12  Later on, the sequence of the first Notch ligands was obtained, and the analysis of the proteins indicated that they all corresponded to cell surface proteins with a large extracellular domain, which further supported a function for Notch as a regulator of cell-cell interactions between neighboring cells. Deciphering the elements downstream of the receptor and ligands involved laborious work from embryologists and biochemists that studied and interpreted the results from different mutant models. Thus, genetic experiments in invertebrates first directed the investigations of the Notch pathway toward the identification of Supressor of Hairless (Su(H)) as a key transducer of neurogenic signals.13,14  Further biochemical studies demonstrated the interaction between Notch and Su(H), the ortholog of the mammalian gene Rbpj (for Recombination-signal Binding Protein jk).15  Genetic screenings were also crucial to identify the Notch-target genes Hairy and Enhancer of Split (Hes), which are HLH (helix loop helix) proteins involved in suppressing the neuronal genes Acute-Scute.16,17  Once again, this connection is conserved in the mammalian systems.

Later on, efforts were focused on investigating Notch signal transduction. The demonstration that Notch together with Su(H)/RBP-J regulated the transcription of Hes genes14,18  provided a strong indication that Notch should function in the nucleus. Indeed, it was initially shown that ectopically expressed intracellular Notch translocated to the nucleus and was capable of activating transcription,19  but ingenious experiments were required to demonstrate the nuclear activity of endogenous Notch.20  Another breakthrough discovery in the history of the Notch field was the demonstration that this receptor was processed by a γ-secretase/presenilin complex in response to ligand binding, and the consequent development of inhibitors that target this activity.21  The usage of these Notch/γ-secretase inhibitors has facilitated the further confirmation of most Notch functions in different cell types and tissues and provided the first attempts of using Notch as a therapeutic target.22,23 

During the last decade, research in the Notch field has been growing exponentially, which has contributed to a better understanding of the relevance of this pathway in the generation and maintenance of multicellular organisms.

Notch is a single-pass transmembrane receptor that can be activated by different transmembrane ligands. In general, Notch binds to one of its ligands located in the adjacent cell, which results in 2 sequential proteolytic cleavages of the receptor. The latter involves the release of the intracellular part of Notch receptor (IC-Notch), its translocation to the nucleus, and its association with RBP-J (also known as CSL for the different ortholog proteins: CBF1, Supressor of Hairless, and Lag1) and the coactivator Mastermind (Mam) to activate specific transcription. Notch participates in the transcriptional regulation of multiple genes, some of them being context-dependent. However, the most important Notch functions have been associated with the regulation of the Hes or Hes-related (Hrt) family of genes. All these genes encode for basic HLH proteins that, in general, function as inhibitors of cell differentiation (Figure 1).

Figure 1

The Notch signaling pathway. Signal-sending Cell: Functional Notch ligands are ubiquitinated by the E3-ubiquitin ligases mindbomb or neuralized. After ligands interact with the Notch receptor, the ligand and the extracellular part of Notch are endocytosed, and ligands may be degraded or recycled. Signal-receiving Cell: The Notch mRNA is translated as a precursor protein, which is cleaved by a furin-like convertase in the Golgi apparatus to produce a functional heterodimeric receptor. During endoplasmic reticulum/Golgi transit, Notch is modified by different glycosyltransferases (Rumi/poglut, pofut, fringes). In cells that express fringe, specific sugar moieties (diamonds) are conjugated to confer higher affinity to Delta-type ligands. After ligand binds to the EGF-like repeats of the Notch extracellular domain, an ADAM metalloprotease cleaves Notch at the S2 site, removing most of the extracellular domain. The membrane-tethered intracellular domain is then cleaved by the Presenilin complex at site S3, either in the plasma membrane or after endocytosis, freeing the Notch intracellular domain (ICN). ICN translocates to the nucleus, displaces the corepressor complex, associates with RBP-J, and recruits coactivators, such as Mastermind. ICN becomes monoubiquitylated (Ub), targeting the receptor for degradation. Several E3 ubiquitin ligases (Deltex, Nedd4, Su(Dx)/Itch, Cbl) can direct Notch receptor trafficking toward lysosomal degradation or toward recycling. Numb can also promote Notch degradation in daughters of an asymmetrically dividing cell.

Figure 1

The Notch signaling pathway. Signal-sending Cell: Functional Notch ligands are ubiquitinated by the E3-ubiquitin ligases mindbomb or neuralized. After ligands interact with the Notch receptor, the ligand and the extracellular part of Notch are endocytosed, and ligands may be degraded or recycled. Signal-receiving Cell: The Notch mRNA is translated as a precursor protein, which is cleaved by a furin-like convertase in the Golgi apparatus to produce a functional heterodimeric receptor. During endoplasmic reticulum/Golgi transit, Notch is modified by different glycosyltransferases (Rumi/poglut, pofut, fringes). In cells that express fringe, specific sugar moieties (diamonds) are conjugated to confer higher affinity to Delta-type ligands. After ligand binds to the EGF-like repeats of the Notch extracellular domain, an ADAM metalloprotease cleaves Notch at the S2 site, removing most of the extracellular domain. The membrane-tethered intracellular domain is then cleaved by the Presenilin complex at site S3, either in the plasma membrane or after endocytosis, freeing the Notch intracellular domain (ICN). ICN translocates to the nucleus, displaces the corepressor complex, associates with RBP-J, and recruits coactivators, such as Mastermind. ICN becomes monoubiquitylated (Ub), targeting the receptor for degradation. Several E3 ubiquitin ligases (Deltex, Nedd4, Su(Dx)/Itch, Cbl) can direct Notch receptor trafficking toward lysosomal degradation or toward recycling. Numb can also promote Notch degradation in daughters of an asymmetrically dividing cell.

Close modal

Notch receptors

There is one Notch receptor in Drosophila, 2 in C elegans and 4 different Notch receptors (Notch1-4) in most vertebrate species, being mammalian Notch1 and Notch2 the most similar to the Drosophila homolog. Notch is a single transmembrane protein composed of an extracellular part with variable number of epidermal growth factor (EGF)-like repeats and an intracellular part containing 7 ankyrin-like repeats, nuclear localization signals, and a transactivation domain. Notch is codified by a single mRNA molecule that translates into a polypeptide, which is cleaved in the Golgi apparatus by a furin-like convertase enzyme.24  This processing generates 2 different fragments (one containing the extracellular domain and another that includes de transmembrane and intracellular domains) that remain associated by disulfide bonds involving a small conserved extracellular domain, LNR (Lin/Notch repeats).25  Hence, the resulting functional Notch is commonly considered as a “heterodimeric” receptor. The LNR region is also crucial to prevent ligand-independent signaling, which is supported by the fact that mutations in this region result in increased Notch activity that associates with T-acute lymphoblastic leukemia.26 

Notch ligands

There are at least 5 functional Notch ligands in vertebrates: 3 orthologs of the Drosophila Delta (Delta or Delta-like [Dll] 1, 3, and 4) and 2 of the Drosophila Serrate (Jagged1 and Jagged2). All ligands are able to interact with the Notch receptor and induce the second cleavage at the extracellular level. However, all ligands have different expression patterns and specific deletion/inhibition of specific ligands results in a very diverse outcome. Notch ligands are also composed of a variable number of EGF-like repeats in their extracellular domain but a small intracellular portion. To achieve Notch activation, Notch ligands need to be internalized in endosomes in the signaling sending cell (before they are presented at the cellular membrane), a process that is regulated through ubiquitination by the E3-ubiquitin ligases Mindbomb and Neuralized.27  The consequence of Mindbomb deficiency is a defective Notch activation, in both invertebrates and vertebrates.28,29 

Notch modification by glycosylation: pofuts, fringes, and pogluts

One of the particularities of Notch is the absence of a downstream signaling cascade. Instead, after Notch activation, IC-Notch travels from the cell surface directly to the nucleus, where it binds RBP-J and indirectly to the DNA. Despite the apparent simplicity of this pathway with no intermediate effectors, Notch signaling involves an extremely accurate regulation, which is multifactorially achieved. For example, multiple enzymes can modify the Notch protein post-translationally, thus changing its functional properties. Pofut-1 is an O-fucosyl-transferase that catalyzes the O-fucosylation of specific EGF-like repeats, which is an essential condition for their subsequent Fringe-dependent modification. Fringe proteins (including Lunatic, Manic, and Radical Fringe) are Golgi-localized glycosyltransferases that add N-acetylglucosamine to O-fucose moieties on EGF-like repeats of the extracellular domains of Notch. Different Fringe homologs modify specific EGF-like repeats with distinct efficiencies.30  Fringe modifications enhance the capacity of Notch to be activated by ligands of the Delta-like family (Dll1,Dll3, and Dll4) but reduce Notch activation by the Serrate/Jagged family of ligands (Jag1-2).31  Although some studies suggest that knockout mutants for all 3 Fringes (in a specific genetic background) do not display more developmental malformations than single mutants for Lunatic Fringe,32  it is well established that different Fringe homologs modulate particular Notch-mediated biologic processes in a specific manner,33-35  such is the case of Lunatic Fringe in the hematopoietic lineage commitment and differentiation.36-39 

Another modification involving the extracellular domain of the Notch receptor is its O-glucosylation mediated by the O-glucosyl-transferase (Poglut), Rumi.40  However, the biochemical effects of this modification are mainly unknown, but it has been suggested that it might regulate the proper folding of Notch that is required for its efficient activation.

Downstream effectors of Notch: the Hes gene family

Hes genes and specifically Hes1 are among the best-characterized Notch target genes. They codify for bHLH proteins and, in general, function as DNA-binding transcriptional repressors. Expression of several members of the family (including Hes1, Hes5, Hes7, Hrt1, and Hrt2) depends mostly on Notch activity and participates in many of the Notch-assigned functions, including proliferation, differentiation, apoptosis, self-renewal, and asymmetric cell division regulation.41 Hes genes are generally responsible for Notch functions that require the inhibition of one specific cell fate to allow the determination of an alternative fate (lateral inhibition), whereas other Notch-inductive functions, such as T-cell specification, may not be dependent on Hes. In this sense, Hes1 is only needed for the first stages of T-cell determination,42  whereas other important Notch targets, such as pTα43  and IL7R,44  are expressed and regulate specific stages of T-cell differentiation.

During embryonic development, specification of the different tissues runs in parallel with the progressive restriction of the stem cell potential. However, significant pools of stem cells are found in the adult tissues that are continuously renewed during lifetime, such as blood or intestine. For many years, HSCs remained the best-characterized somatic stem cells and, consequently, they have been used as a model to study adult tissue regeneration. In recent years, similar types of cells have been identified in many other tissues, such as muscle, neuronal, pancreas, lung, and breast, among others, and it is now widely accepted that they are crucial players in tissue homeostasis and regeneration. In the blood system, there is now definitive evidence that the somatic-, blood-specific stem cells are originated during embryonic life and maintained thereafter. Although Notch is generally considered as an essential signaling pathway that regulates multiple stem cell functions, its participation diverges among the different organs and tissues. For example, in the mammalian hematopoietic system, Notch1 is required for the generation of HSCs in the embryo, but it is dispensable for the maintenance of the HSCs in the adult organism,45  whereas the neuronal stem cells depend on Notch in both embryonic and adult life. Our current understanding on how Notch contributes to tissue homeostasis includes a plethora of functions in both the stem and progenitor cell populations that cannot be generalized from one tissue to the others. In this review, we summarize what is known about Notch functions during generation and maintenance of HSCs and compare this with its role in other tissue-specific stem cell populations.

Multiple types of specialized cells, which are responsible for nutrient transport and immune defense, constitute the hematopoietic system. During the adult life, all hematopoietic cells in the organism are renewed every 5 to 120 days depending on the cell type. Continuous production of limited numbers of blood cells is achieved by the differentiation of HSCs in the bone marrow microenvironment. However, it is the exclusive capacity of HSCs for self-renewal that guarantees the maintenance of this tissue throughout life. The question that arises here is as follows: how this rare population acquires the features of self-renewal and pluripotency that define a stem cell. From our knowledge, stem cell properties are acquired during embryonic life; thus, in the following sections, we review what it is known about the mechanisms that regulate the generation and maintenance of HSCs and the role of Notch in these different processes.

Embryonic hematopoiesis

Before colonizing the bone marrow, newly formed HSCs as well as all different hematopoietic lineages are found in the developing vertebrates in a tightly regulated spatial and temporal manner. More specifically, embryonic hematopoiesis takes place in 2 different waves (ie, primitive and permanent-definitive hematopoiesis).46  In mammals, primitive hematopoiesis occurs primarily in the blood islands of the yolk sac47,48  around murine embryonic day 7.5 (E7.5),49  and whether yolk sac blood cells originate from a common endothelial progenitor called hemangioblast50,51  is still unclear.48  Grafting experiments using quail-chick chimeras and colony-forming unit cultures showed that the yolk sac mainly produces primitive erythrocytes and myeloid progenitors but does not generate definitive HSCs.52-56  Indeed, HSCs are first detected in the yolk sac after circulation between the yolk sac and the embryo is established (in the mouse embryo, HSCs are found at E9 and circulation starts around E8.5).57  Placenta is also an important source of erythroid and myeloid progenitors at E9.0; and interestingly, it becomes a reservoir of HSCs around E12.58-60  However, the first definitive HSCs considering those cells with the capacity to reconstitute irradiated adult mice appear in the dorsal aorta, in a region surrounded by gonad and mesonephros (also known as the AGM region) around day 10 of murine development,61,62  closely associated with the endothelial cells (Figure 2A). Cell-tracing experiments demonstrated that murine adult hematopoietic cells originate from embryonic cells that express VE-cadherin and Runx1 around E9.0, strongly suggesting the endothelial embryonic origin of HSCs but not excluding other possibilities.63,64  Importantly, endothelial cells from fetal liver are no longer capable of generating hematopoietic cells, indicating that a putative common endothelial-hematopoietic progenitor should be very restricted both temporally and spatially to specific embryonic vessels, such as the dorsal aorta.65 

Notch in embryonic arterial and hematopoietic development

One of the best-characterized Notch functions in vertebrates consists of the determination of the arterial program, and several Notch mutant embryos in both zebrafish and mouse lack artery specification as determined by the absence of EphrinB2, CD44, or SMA expression.66-68  Accordingly, because HSCs originate in arterial vessels, the predicted phenotype of Notch mutant embryos was indeed lack of hematopoiesis. This is the phenotype of the Notch1-, Rbpj-, and Mindbomb-deficient mutant mice.69-71  In contrast, coup-TFII mutants show ectopic Notch activation in veins accompanied by the expression of arterial markers and ectopic hematopoiesis.72  Other mutants that fail to define the artery-vein boundaries, such as Activin A-receptor type II-like1 (acvrl1, alk1) or endoglin (CD150), show ectopic hematopoiesis in veins or at least in vein-like vessels that do not express the arterial marker EphrinB2.72-74  This link between arterial specification and hematopoiesis has complicated a direct assignment of the hematopoietic defects to Notch deficiency because secondary effects of arterial development may result in the same phenotype. Opportunely, experiments in zebrafish showed that ectopic Notch1 activation in the veins induced specific hematopoietic gene expression, including Runx1 and c-myb, in the absence of arterial differentiation.71  However, a formal proof that HSCs can be generated in the absence of arteries is still missing. Further insight into the question on how Notch regulates hematopoiesis independently of its role on arterial specification was obtained by the analysis of the Jagged1 mutant. In this work, we demonstrated that Jagged1, Jagged2, and Delta4 are all expressed in the endothelium of the aorta in the developing AGM.70  However, analysis of the mutants demonstrated that they do not display redundant functions. Specifically, Delta4-deficient mice show a strong arterial phenotype67,68  and die at early developmental time. Similarly, Jagged1 mutants show some vascular malformations75  but still express arterial markers, such as EphrinB2, CD44, and SMA, and importantly they mostly failed to generate hematopoietic cells.76  These results undoubtedly demonstrated that Notch was required for definitive hematopoiesis in vivo, and uncoupled this Notch function from its role in arterial specification.

Another unexpected result that arose from the analysis of mutant mice was the fact that only definitive hematopoiesis in the AGM region, but not the embryonic hematopoiesis of the yolk sac, was Notch-dependent. As discussed in “Embryonic hematopoieisis” murine yolk sac from E7.5 to E8.5 mainly produces primitive nucleated erythrocytes (an event known as primitive erythropoiesis), and it is not until E9.0 when circulation is already established that the yolk sac contains different hematotopoietic progenitors capable of generating myeloid, erythroid, or lineage-mixed colonies in culture, but also definitive HSCs that are transplantable and able to reconstitute busulphan-treated newborn mice. Importantly, the yolk sac from Notch-deficient mutants contains similar numbers of hematopoietic progenitors at different times of development compared with their wild-type or heterozygous counterparts69,77  but lacks the HSC potential at E9.0 (15-20 somites).69  Together, these results indicate that Notch signaling is dispensable for primitive hematopoiesis, whereas, independently of the site of origin (provided that there is more than 1), Notch signaling is essential for generating definitive hematopoiesis. In agreement with this idea, analysis of chimeric mice generated from wild-type and Notch1-deficient ES cells demonstrated that Notch1-deficient cells contribute to all types of embryonic, fetal progenitors and hematopoietic cells present in fetal liver but do not contribute to adult hematopoiesis.78  Similarly, mutation of Mindbomb in zebrafish affected the later wave of hematopoietic cells (characterized by expression of c-myb or Runx1) but not for the early phase of primitive cells.79 

Notch in fetal hematopoiesis

The final purpose of developing HSCs is to access their niche in the bone marrow that will allow HSC self-renewal and differentiation throughout the adult life. However, before reaching the bone marrow, newly formed HSCs migrate to the fetal liver, which is the main hematopoietic organ before birth. During this period, early lymphoid fetal liver progenitors need to colonize the thymic epithelial primordium where they experienced high Notch signaling dose and differentiate into T cells,80  whereas HSCs will need to colonize the bone marrow. However, the most remarkable hematopoietic process in the fetal liver is that HSCs increase in number. For this reason, fetal liver signals have been studied for many years in an attempt to identify candidate molecules that prone HSC expansion. Whether Notch participates in this particular fetal process is mainly unknown basically because most of conventional Notch mutants are either lethal or failed to generate hematopoiesis before the fetal liver stage. Thus, analysis of conditional Notch mutants that specifically affect the fetal liver stage may be useful to uncover any possible role of Notch in this process.

Notch in adult HSCs

Early studies demonstrated that Notch is expressed in the undifferentiated progenitors from human bone marrow cells81  and suggested a putative role for this factor in leukemia.10  Later on, experiments using myeloid progenitor cell lines indicated that Notch is an essential regulator of hematopoietic differentiation,82,83  thus opening the possibility that it might function in preserving the stem cell phenotype. In addition, parathyroid hormone, which is a crucial regulator of the osteoblastic HSC niche in the bone marrow, increases the levels of Jagged1 in this tissue with in vitro evidence of Notch function in HSC expansion.84  However, the analysis of transgenic mice carrying a dominant negative form of the coactivator Mastermind (which specifically blocks all canonical Notch signaling) or mice deficient for Rbpj indicated that Notch activity was dispensable for the maintenance of HSCs in the adult bone marrow under physiologic conditions.85  Further support for these results can be found in the conditional deletion of Notch1 or Notch1 plus Notch2 under the control of the interferon-dependent expression of Mx-Cre that specifically and exclusively affected lymphoid differentiation, but not other hematopoietic lineages.86-88  Consistent with the notion that Notch activity is dispensable in the adult HSCs, a recently published database (www.immgen.org)89  showed low levels of different Notch receptors in purified HSCs.

Notch-dose dependency in hematopoietic differentiation and hematopoietic disorders

Experimental systems that mostly rely on complete abrogation or strong persistent activation of the Notch pathway result in phenotypes that are mainly associated with the lymphoid lineage consistent with the fact that high Notch activity promotes T-cell differentiation, whereas complete absence of Notch induces early B-cell commitment.86,90  In agreement with this, activating Notch mutations are commonly found in T-cell leukemias.91,92  However, whole genome sequencing of B-chronic lymphocytic leukemia cells has revealed that 12% of these samples also contain activating mutations of Notch associated with a Notch-active signature,93  suggestive of a regulatory role for Notch in B-cell differentiation, which is also in agreement with the effects of Rbpj or Notch2 deletion in committed B-cell progenitors that impaired marginal zone B cells and potentiates follicular B-cell differentiation in the spleen.94,95  Unexpectedly, the identified activating mutations in B-chronic lymphocytic leukemia affect the Notch1 receptor, which is not the main effector for B-cell terminal differentiation, suggesting that aberrant transforming Notch activity can be delivered by any receptor. Indeed, this emphasizes the importance of large-scale genome sequencing of patients with different hematopoietic disorders in providing a new perspective about the involvement of Notch in these malignancies. The knowledge on how much, when,and where Notch is required to maintain the correct specification of the hematopoietic lineages (including the HSCs) is an essential requirement for further manipulation of the Notch pathway for biomedical purposes.

However, rather than just being a Notch ON/OFF issue, the extent of Notch activity is also critical for most of the Notch functions involved in the homeostasis of the hematopoietic lineages. Alternative approaches that overcome this limitation include the intervention on positive or negative regulators, which might affect the strength of Notch signal. Indeed, overexpression of Lunatic fringe that reduces the affinity between Notch and Delta ligands forces lymphoid progenitors to become B cells in a high Delta-presenting environment,39  whereas modulating the density of Delta1 influences the generation of B or T cells in vitro.96  Recent works describing the phenotype associated with loss-of-function mutations of positive Notch regulators, such as Mindbomb,97  Nicastrin,98  Presenilin,99  or Pofut,100  suggested that reduction in the levels of Notch activity is sufficient to induce a myeloproliferative syndrome that phenocopies the effects observed in one of the Mx-Cre-mediated Notch1 and Notch2 deletion,98  despite that the same experimental model was previously generated showing an exclusive lymphoid phenotype.88  Consistent with the possibility that myeloid differentiation/proliferation is favored by reduced Notch activity (see model in Figure 2), loss-of-function mutations of the pathway are present in approximately 12% of patients with chronic myelomonocytic leukemia.98  Interestingly, mouse transplanted with hematopoietic cells in which RBPJ-mediated Notch activity is completely abrogated, including Rbpj-deletion or ectopic DN-Mam expression,85  did not show any myeloproliferative phenotype, suggesting that RBPJ-independent or non–cell-autonomous Notch activities are required to develop the disease. In addition, a different study showed that expression of DN-Mam precludes megakaryocyte differentiation in vivo.101 

Figure 2

Pleiotropic Notch functions in the hematopoietic system. (A) Notch is required during hematopoietic development to generate HSCs in the AGM. This process takes place in the aorta endothelium and is dependent on the Jagged1 ligand and the Notch1 receptor. Endothelial-like pre-HSCs or HSCs are the putative target of Notch activation (light green). (B-C) Models of Notch signaling in hematopoietic differentiation. (B) Notch is involved at different steps of differentiation of the hematopoietic lineages. The classic hierarchical model of the hematopoietic tree is here adapted to the concept that a gradient of Notch activation or signaling determines the different types of hematopoietic cells. (C) A hematopoietic potential restriction model107  in which lineage restriction is determined by Notch doses. HSCs progressively lose their capacity to generate different lineages concomitantly to different exposure to Notch doses. Intensity of the blue color represents a speculative gradient of Notch activity. Dashed lines represent cell potential to become a specific cell type. Solid lines represent progression through normal hematopoietic development. LT-HSC indicates long-term HSCs; CMP, common myeloid progenitor; LMPP, lymphoid/myeloid potential progenitor; CME, common megakaryocyte-erythroid progenitor; ETP, early T-progenitor; Ery, erythrocyte; Meg, megakaryocyte; and M/Gr, macrophage/granulocyte.

Figure 2

Pleiotropic Notch functions in the hematopoietic system. (A) Notch is required during hematopoietic development to generate HSCs in the AGM. This process takes place in the aorta endothelium and is dependent on the Jagged1 ligand and the Notch1 receptor. Endothelial-like pre-HSCs or HSCs are the putative target of Notch activation (light green). (B-C) Models of Notch signaling in hematopoietic differentiation. (B) Notch is involved at different steps of differentiation of the hematopoietic lineages. The classic hierarchical model of the hematopoietic tree is here adapted to the concept that a gradient of Notch activation or signaling determines the different types of hematopoietic cells. (C) A hematopoietic potential restriction model107  in which lineage restriction is determined by Notch doses. HSCs progressively lose their capacity to generate different lineages concomitantly to different exposure to Notch doses. Intensity of the blue color represents a speculative gradient of Notch activity. Dashed lines represent cell potential to become a specific cell type. Solid lines represent progression through normal hematopoietic development. LT-HSC indicates long-term HSCs; CMP, common myeloid progenitor; LMPP, lymphoid/myeloid potential progenitor; CME, common megakaryocyte-erythroid progenitor; ETP, early T-progenitor; Ery, erythrocyte; Meg, megakaryocyte; and M/Gr, macrophage/granulocyte.

Close modal

On the other hand, most of the phenotypes associated with Notch pathway manipulation suggest an almost exclusive cell-autonomous requirement for Notch signaling in the maintenance of the hematopoietic system. However, mice deficient for Pofut1, which is required upstream of Fringe glycosyltransferases to permit Notch signaling through Delta, displayed both cell-autonomous and non–cell-autonomous defects that resulted in myeloproliferative disease,100  suggestive of undetermined Notch functions in the regulation of the stromal HSC niche. In addition, defective Notch signaling in the murine skin results in an epidermal-barrier-associated inflammatory phenotype, which leads to massive thymic stroma lymphopoietin (TSLP) expression and produces lethal B-cell102  or myeloid proliferative disorders.103 

In conclusion, and to help understand how Notch impinges in the hierarchy of hematopoietic lineages, we propose a Notch-dose-based model that is compatible with recent data and revised models104-107  in which specific Notch activity levels impose progressive lineage restrictions (see model in Figure 2B-C).

Notch in ex vivo hematopoietic cultures and stress conditions

Nowadays there is no evidence for a physiologic role of Notch in the maintenance of HSCs in vivo. Despite this fact, Bernstein's laboratory has recently demonstrated that Notch pathway manipulation is a potential way of expanding HSCs or progenitor cells for clinical applications.108-110  The basis for this finding comes from early experiments in which murine undifferentiated bone marrow cells (LinSca+Kit+) were transduced with retrovirus encoding active Notch1. In this pioneer experiment, cells with self-renewal and pluripotent myeloid and lymphoid differentiation capacity in vitro and in vivo were obtained.108  To avoid using retrovirally infected cells, they have now optimized the culture conditions to activate endogenous Notch through immobilized Notch ligands.109  This strategy turned out to be particularly efficient for the expansion of short-term lymphoid and myeloid progenitors from samples with low numbers of these cell populations, such as cord blood. This strategy is now under clinical investigation in immune-ablated patients, which are being transplanted with one unmanipulated plus one Notch-expanded cord blood unit. The first evaluated patients have significantly reduced the time of neutrophil engraftment compared with patients transplanted with 2 unmanipulated cord blood units.110  However, Notch-dependent expansion protocols can still be improved to enhance the long-term engraftment of the manipulated unit after the identification of crucial elements provided by endothelial cells111  or using different ligands.112  At the mechanistic level, the impact of Notch activation on ex vivo expansion of long-term or short-term HSCs may be directly correlated with its capacity to regulate cell cycle in HSCs113  or/and hematopoietic recovery under stress conditions.87 

Different Notch functions that take place in particular cell types, tissues, or developmental stages involve specific mechanistic strategies that are under intense investigation. For example, many efforts have been made to understand how Notch regulates stem cell maintenance and cell fate determination in both embryonic and regenerating adult tissues. Despite that some of the identified mechanistic strategies can be tissue specific, others maybe not, or in any case they can serve as inspiration for people working on different systems (Figure 3). In the next section, we have selected some of these examples, which may apply to the hematopoietic system.

Figure 3

Notch-dependent mechanistic strategies for stem cell maintenance and differentiation. (A) Neuronal differentiation in the developing neuroepithelium is dependent on an asymmetric cell division in which Numb is asymmetrically distributed and inhibits Notch signaling in one of the cells to permit neuronal differentiation. (B) In the developing brain, Notch controls the oscillation of downstream genes (hes1 and Ngn2) that allows controlled proliferation and differentiation waves in the midbrain compartment. In the boundary (green boxes), Hes1 protein levels are sustained, Ngn2 levels are low, and proliferation and neuron formation are inhibited.141  (C) Notch promotes cell proliferation in the basal layer of the embryonic epidermis, however, in adult epidermis. (D) Notch is inhibiting proliferation and inducing differentiation of the interfollicular basal layer. In the bulge, Notch and Jag1 are regulating epidermal stem cells by promoting the follicular stem cell fate downstream of Wnt/β-catenin. (E) Two types of ISCs have been identified: Lgr5+ proliferating cells and quiescent +4 cells (Bmi+). Lgr5+ cells are located at the bottom of the intestinal crypts, next to an intestinal secretory-type cell called Paneth cell. These cells are crucial ISC-niche elements, express Notch-ligands Dll1 and Dll4, and are required to maintain ISCs (probably through Notch activation). However, differentiation to the secretory lineage, including Paneth cells, requires Atonal, which is a target of Notch/hes1 repression, suggesting that generation of the ISC niche is under the control of ISCs through Notch. (F) Notch signaling induced by Delta1 is required for muscle regeneration on injury. The balance between Notch and TGFb/smad3 signals determines the levels of CDK inhibitors and the proliferative capacity of muscle progenitors.

Figure 3

Notch-dependent mechanistic strategies for stem cell maintenance and differentiation. (A) Neuronal differentiation in the developing neuroepithelium is dependent on an asymmetric cell division in which Numb is asymmetrically distributed and inhibits Notch signaling in one of the cells to permit neuronal differentiation. (B) In the developing brain, Notch controls the oscillation of downstream genes (hes1 and Ngn2) that allows controlled proliferation and differentiation waves in the midbrain compartment. In the boundary (green boxes), Hes1 protein levels are sustained, Ngn2 levels are low, and proliferation and neuron formation are inhibited.141  (C) Notch promotes cell proliferation in the basal layer of the embryonic epidermis, however, in adult epidermis. (D) Notch is inhibiting proliferation and inducing differentiation of the interfollicular basal layer. In the bulge, Notch and Jag1 are regulating epidermal stem cells by promoting the follicular stem cell fate downstream of Wnt/β-catenin. (E) Two types of ISCs have been identified: Lgr5+ proliferating cells and quiescent +4 cells (Bmi+). Lgr5+ cells are located at the bottom of the intestinal crypts, next to an intestinal secretory-type cell called Paneth cell. These cells are crucial ISC-niche elements, express Notch-ligands Dll1 and Dll4, and are required to maintain ISCs (probably through Notch activation). However, differentiation to the secretory lineage, including Paneth cells, requires Atonal, which is a target of Notch/hes1 repression, suggesting that generation of the ISC niche is under the control of ISCs through Notch. (F) Notch signaling induced by Delta1 is required for muscle regeneration on injury. The balance between Notch and TGFb/smad3 signals determines the levels of CDK inhibitors and the proliferative capacity of muscle progenitors.

Close modal

The oscillation model: neural stem cells

Similar to HSCs, neural stem cell generation also occurs during embryonic life. However, some specific regions of the adult hippocampus (subgranular zone), lateral ventricle (subventricular zone), and the olfactory bulb still contain some functional neural cells with self-renewal capacity. During embryonic development, different phases of both symmetric and asymmetric cell divisions are required to generate all main neuron lineages: First, symmetric divisions of the neural stem cells support the expansion of the neuroepithelial progenitors and the generation of radial glial cells. Next, asymmetric division of these progenitors gives rise to all types of cells during the neurogenic and gliogenic phases. It is well established that Notch pathway is a crucial regulator of this process, and it is required to maintain neuronal progenitors and inhibit neuronal differentiation.114  A nice example on how Notch is differentially activated in the developing neurons is found during asymmetric division of cortical neuronal progenitors. In this process, both the Notch receptor and its inhibitor Numb are asymmetrically distributed in the 2 daughter cells. The one carrying Notch induces hes1 transcription, which directly inhibits neuronal genes. However, the system is not so simple because Hes1 protein inhibits its own transcription, thus generating a negative feedback loop that creates an oscillatory pattern of hes1 expression.115  Oscillations on Hes1 protein levels induce the oscillation of its target genes, including Neurogenin2 (Ngn2), but also Delta1 (Dll1), which in turn induces oscillation of Notch activity in neighboring cells.116  Elegant studies with time-lapse imaging from Kageyama's laboratory showed that Ngn2 and Dll1 oscillate in neural stem/progenitors in an opposite phase as hes1, and their expression is sustained in differentiating neurons where Hes1 expression is repressed.117  This oscillatory system is responsible for maintaining active proliferation and neuronal determination in the developing midbrain, whereas sustained Hes1 levels in the cells of the boundary of the forebrain, midbrain, and hindbrain maintain low levels of Ngn2 that result in low proliferation and prevent neuronal formation.

In the adult brain, neurogenesis is much more restricted in the number and differentiation potential of cells undergoing this process. However, Notch seems to be also involved in the maintenance of the adult neural stem cell population, whereas inactivation of Notch signaling induces neuronal differentiation and depletes the neural stem cell pool.118,119 

Cell- versus non–cell-autonomous or embryonic versus adult Notch effects: skin stem cells

The adult skin epidermis contains 2 different types of progenitor or stem cells: the proliferative cell compartment, which is located in the basal layer of the interfolicullar skin and can differentiate into a single lineage; and the mostly quiescent multipotent stem cell compartment, restricted to the bulge of the hair follicle. The bulge stem cells (which are the pluripotent skin stem cells) can generate both epidermal and hair follicle cells dependent on Notch activity because Notch-RPP-J signaling promotes the follicular fate.120,121  The activation of Notch in this system is downstream of β-catenin activity through the transcriptional activation of Jagged1.122  However, there is no evidence for Notch function in maintaining the multipotent bulge stem cell compartment.

In the basal epidermal layer, Notch signaling regulates proliferation and differentiation of the epidermal cells; however, some data are still controversial and mainly depend on the experimental systems used. Early studies using epidermis-specific Notch1 knockout mice (using Keratin5-Cre) demonstrated that Notch deletion increased skin proliferation by down-regulating p21 in the keratinocytes,123  thus creating a tumor-promoting environment with increased β-catenin levels.124  These findings, together with data from knockdown and knockout of different Notch elements in the mouse skin, supported the idea that Notch behaved as a tumor suppressor in this tissue.125  Surprisingly, Rbpj deletion in the developing skin (using the keratin14-CRE mouse line) resulted in a hypoproliferative phenotype concomitant with a blockage in skin differentiation that was non–cell-autonomous because it was recovered when mutant skin was grafted onto nude mice.126  These results, further than creating controversy, highlighted the great complexity of the interactions that mediate Notch effects and suggested that non–cell-autonomous effects might also influence the effect of Notch in skin tumorigenesis. Indeed, using a mouse model with a chimeric pattern of Notch1 deletion in the skin, it was recently demonstrated that it is not the absence of Notch in the keratinocyte progenitors but the skin barrier defects induced by Notch deletion, that is responsible for tumor promotion in these mutants.127  Analysis of mice that are mutant for the Notch-processing enzymes γ-secretase128  and Adam10129  have further confirmed that Notch plays both cell-autonomous and non–cell-autonomous effects in the adult skin. In the embryo, deletion of Rbpj prevented the transition between basal to spinous layers,126  whereas Adam10 mutants showed a premature differentiation from spinous to granular layers,129  which in both cases generates an aberrantly thin spinous layer. Of note, embryonic inactivation of γ-secretase did not impose any phenotype to the developing skin.

The niche-forming stem cell model: intestinal stem cells

The intestinal epithelium is one of the most rapidly self-renewing tissues of the organism (every 4-5 days for most cell types). In the adult, all postmitotic cell lineages are originated by terminal differentiation of a progenitor pool derived from the intestinal stem cells (ISCs). A few years ago, it was proposed that a highly proliferative population of cells residing in the bottom of the crypts, characterized by expression of the Lgr5 marker, were the actual ISCs. Importantly, these cells reside adjacent to terminally differentiated Paneth cells that support their self-renewal and pluripotent capacities by providing Wnt and Notch ligands.130 

Recently, it has been shown that Lgr5+ cells can derive from quiescent Bmi1 expressing cells located in the +4 position (relative to the bottom of the crypt),131  which suggest the coexistence of more than one ISCs compartment.132  Notch plays an essential role in the maintenance of intestinal homeostasis because inhibiting Notch, both pharmacologically and genetically, impairs the integrity of the intestinal crypt leading to the accumulation of postmitotic goblet (mucus-secreting) cells.22,133  However, this effect is supposed to be dependent on Hes1, which inhibits Atonal that is required for differentiation into the secretory lineage.134  Notch is also expressed in the stem cell compartment of the intestine,135  and mice carrying specific intestinal deletion of Delta1 to 4 ligands failed to activate Notch, lacking the expression of stem cell markers in the crypt cells (including Lgr5 and Olfm4).136  This fact suggests a role for Notch, not only in regulating intestinal differentiation but also in the maintenance of the ISCs. However, the fact that secretory Paneth cells (that depends on Atonal, which is inhibited downstream of Notch) participate in the intestinal stem cell niche precludes giving a definitive answer about whether Notch directly regulates intestinal stemness or is just an indirect effect because of possible Paneth cell number alterations. However, in a simplistic viewpoint, Notch inhibition should lead to increased Paneth cell numbers and favors ISCs generation, which is not the case.

The role of Notch in regulating the niche for stem cell generation or maintenance is a novel concept, with emerging evidence, not only in the mammalian intestinal system but also in the development of the Drosophila gut137  as well as the hematopoietic development of zebrafish,138  which should be investigated in the near future.

A regenerative medicine hallmark: myogenesis

Finally, another important function of Notch is as a regulator of myogenesis both during embryonic somitogenesis and in postnatal muscle repair. Importantly, inducing Notch signaling by Delta1 potentiate muscle regeneration in old aged mice. Specifically, the balance between Notch and TGFβ/smad3 signals determines the levels of CDK inhibitors that are crucial regulators of the proliferative capacity of muscle progenitors.139  Similarly, activation of Notch in satellite cells blocks myogenesis, whereas inhibition of the pathway leads to increased cell differentiation, indicating that Notch signal is essential to regulate the pool of satellite cells and adult tissue repair.140 

In conclusion, one of the take-home messages of this review is the essential role of Notch in the regulation of most binary cellular decisions that take place both in the embryo and in adult-regenerating tissues, which is exemplified in the hematopoietic system. It is thus envisioned that the possibility to modulate Notch signaling will open an avenue for regenerative medicine applications, including hematologic and nonhematologic diseases, but also for cancer treatment. Before that, much more detailed studies, including mechanistic data from hematopoiesis but also other embryonic and adult tissues, are required to clearly determine whether and how Notch can be used as a tool for generating clinically relevant and safe cell populations for transplantation.

The authors thank the members of the Bigas-Espinosa laboratory for critical discussions, especially Teresa D'Altri, Leonor Norton, Julia Inglés-Esteve, and Erika López-Arribillaga for critical reading of the manuscript.

The authors apologize to those whose work could not be cited because of space limitation.

This work was supported by Ministerio Ciencia e Innovación (SAF2007-60080, PLE2009-0111, SAF2010-15450, PI10/01128), Red Temática de Investigación Cooperativa en Cáncer (RD06/0020/0098), and Agència de Gestió d'Ajuds Universitaris i de Recerca (AGAUR; 2009SGR-23 and CONES2010-0006). L.E. is an investigator of ISCsIII program (02/30279).

Contribution: L.E. and A.B. wrote the review.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Anna Bigas, Institut Mar Investigacions Mèdiques, Hospital del Mar, Dr Aiguader 88, 08003 Barcelona, Spain; e-mail: abigas@imim.es.

1
Artavanis-Tsakonas
 
S
Rand
 
MD
Lake
 
RJ
Notch signaling: cell fate control and signal integration in development.
Science
1999
, vol. 
284
 
5415
(pg. 
770
-
776
)
2
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
, vol. 
92
 (pg. 
367
-
409
)
3
Morgan
 
TH
The theory of the gene.
American Naturalist
1917
, vol. 
51
 
609
pg. 
513
 
4
Artavanis-Tsakonas
 
S
Muskavitch
 
MA
Yedvobnick
 
B
Molecular cloning of Notch, a locus affecting neurogenesis in Drosophila melanogaster.
Proc Natl Acad Sci U S A
1983
, vol. 
80
 (pg. 
1977
-
1981
)
5
Kidd
 
S
Lockett
 
TJ
Young
 
MW
The Notch locus of Drosophila melanogaster.
Cell
1983
, vol. 
34
 
2
(pg. 
421
-
433
)
6
Yochem
 
J
Weston
 
K
Greenwald
 
I
The Caenorhabditis elegans lin-12 gene encodes a transmembrane protein with overall similarity to drosophila notch.
Nature
1988
, vol. 
335
 
6190
(pg. 
547
-
550
)
7
Yochem
 
J
Greenwald
 
I
glp-1 and lin-12, genes implicated in distinct cell-cell interactions in C. elegans, encode similar transmembrane proteins.
Cell
1989
, vol. 
58
 
3
(pg. 
553
-
563
)
8
Coffman
 
C
Harris
 
W
Kintner
 
C
Xotch, the Xenopus homolog of Drosophila notch.
Science
1990
, vol. 
249
 (pg. 
1438
-
1441
)
9
del Amo
 
FF
Smith
 
DE
Swiatek
 
PJ
, et al. 
Expression pattern of Motch, a mouse homolog of Drosophila Notch, suggests an important role in early postimplantation mouse development.
Development
1992
, vol. 
115
 
3
(pg. 
737
-
744
)
10
Ellisen
 
LW
Bird
 
J
West
 
DC
, et al. 
TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms.
Cell
1991
, vol. 
66
 (pg. 
649
-
661
)
11
Seydoux
 
G
Greenwald
 
I
Cell autonomy of lin-12 function in a cell fate decision in C. elegans.
Cell
1989
, vol. 
57
 (pg. 
1237
-
1245
)
12
Wharton
 
KA
Johansen
 
KM
Xu
 
T
Artavanis-Tsakonas
 
S
Nucleotide sequence from the neurogenic locus notch implies a gene product that shares homology with proteins containing EGF-like repeats.
Cell
1985
, vol. 
43
 
3
(pg. 
567
-
581
)
13
Schweisguth
 
F
Posakony
 
JW
Suppressor of Hairless, the Drosophila homolog of the mouse recombination signal-binding protein gene, controls sensory organ cell fates.
Cell
1992
, vol. 
69
 
7
(pg. 
1199
-
1212
)
14
Fortini
 
ME
Artavanis-Tsakonas
 
S
The suppressor of hairless protein participates in notch receptor signaling.
Cell
1994
, vol. 
79
 
2
(pg. 
273
-
282
)
15
Matsunami
 
N
Hamaguchi
 
Y
Yamamoto
 
Y
, et al. 
A protein binding to the J kappa recombination sequence of immunoglobulin genes contains a sequence related to the integrase motif.
Nature
1989
, vol. 
342
 
6252
(pg. 
934
-
937
)
16
Bailey
 
AM
Posakony
 
JW
Suppressor of Hairless directly activates transcription of Enhancer of Split complex genes in response to Notch receptor activity.
Genes Dev
1995
, vol. 
9
 
21
(pg. 
2609
-
2622
)
17
Vassin
 
H
Vielmetter
 
J
Campos-Ortega
 
JA
Genetic interactions in early neurogenesis of Drosophila melanogaster.
J Neurogenet
1985
, vol. 
2
 
5
(pg. 
291
-
308
)
18
Jarriault
 
S
Brou
 
C
Logeat
 
F
Schroeter
 
EH
Kopan
 
R
Israel
 
A
Signalling downstream of activated mammalian Notch.
Nature
1995
, vol. 
377
 
6547
(pg. 
355
-
358
)
19
Fortini
 
ME
Rebay
 
I
Caron
 
LA
Artavanis-Tsakonas
 
S
An activated notch receptor blocks cell-fate commitment in the developing drosophila eye.
Nature
1993
, vol. 
365
 
6446
(pg. 
555
-
557
)
20
Schroeter
 
EH
Kisslinger
 
JA
Kopan
 
R
Notch-1 signalling requires ligand-induced proteolytic release of intracellular domain.
Nature
1998
, vol. 
393
 
6683
(pg. 
382
-
386
)
21
De Strooper
 
B
Annaert
 
W
Cupers
 
P
, et al. 
A presenilin-1-dependent gamma-secretase-like protease mediates release of Notch intracellular domain.
Nature
1999
, vol. 
398
 
6727
(pg. 
518
-
522
)
22
van Es
 
JH
van Gijn
 
ME
Riccio
 
O
, et al. 
Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells.
Nature
2005
, vol. 
435
 
7044
(pg. 
959
-
963
)
23
Real
 
PJ
Tosello
 
V
Palomero
 
T
, et al. 
Gamma-secretase inhibitors reverse glucocorticoid resistance in T cell acute lymphoblastic leukemia.
Nat Med
2009
, vol. 
15
 
1
(pg. 
50
-
58
)
24
Logeat
 
F
Bessia
 
C
Brou
 
C
, et al. 
The Notch1 receptor is cleaved constitutively by a furin-like convertase.
Proc Natl Acad Sci U S A
1998
, vol. 
95
 
7
(pg. 
8108
-
8112
)
25
Blaumueller
 
CM
Qi
 
H
Zagouras
 
P
Artavanis Tsakonas
 
S
Intracellular cleavage of notch leads to a heterodimeric receptor on the plasma membrane.
Cell
1997
, vol. 
90
 
2
(pg. 
281
-
291
)
26
Weng
 
AP
Ferrando
 
AA
Lee
 
W
, et al. 
Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia.
Science
2004
, vol. 
306
 
5694
(pg. 
269
-
271
)
27
Le Borgne
 
R
Remaud
 
S
Hamel
 
S
Schweisguth
 
F
Two distinct E3 ubiquitin ligases have complementary functions in the regulation of delta and serrate signaling in Drosophila.
PLoS Biol
2005
, vol. 
3
 
4
pg. 
e96
 
28
Itoh
 
M
Kim
 
CH
Palardy
 
G
, et al. 
Mind bomb is a ubiquitin ligase that is essential for efficient activation of Notch signaling by Delta.
Dev Cell
2003
, vol. 
4
 
1
(pg. 
67
-
82
)
29
Koo
 
BK
Lim
 
HS
Song
 
R
, et al. 
Mind bomb 1 is essential for generating functional Notch ligands to activate Notch.
Development
2005
, vol. 
132
 
15
(pg. 
3459
-
3470
)
30
Wang
 
Y
Shao
 
L
Shi
 
S
, et al. 
Modification of epidermal growth factor-like repeats with O-fucose: molecular cloning and expression of a novel GDP-fucose protein O-fucosyltransferase.
J Biol Chem
2001
, vol. 
276
 
43
(pg. 
40338
-
40345
)
31
Yang
 
LT
Nichols
 
JT
Yao
 
C
Manilay
 
JO
Robey
 
EA
Weinmaster
 
G
Fringe glycosyltransferases differentially modulate Notch1 proteolysis induced by Delta1 and Jagged1.
Mol Biol Cell
2005
, vol. 
16
 
2
(pg. 
927
-
942
)
32
Moran
 
JL
Shifley
 
ET
Levorse
 
JM
, et al. 
Manic fringe is not required for embryonic development, and fringe family members do not exhibit redundant functions in the axial skeleton, limb, or hindbrain.
Dev Dyn
2009
, vol. 
238
 
7
(pg. 
1803
-
1812
)
33
Benedito
 
R
Roca
 
C
Sorensen
 
I
, et al. 
The notch ligands Dll4 and Jagged1 have opposing effects on angiogenesis.
Cell
2009
, vol. 
137
 
6
(pg. 
1124
-
1135
)
34
Evrard
 
YA
Lun
 
Y
Aulehla
 
A
Gan
 
L
Johnson
 
RL
Lunatic fringe is an essential mediator of somite segmentation and patterning.
Nature
1998
, vol. 
394
 
6691
(pg. 
377
-
381
)
35
Xu
 
K
Nieuwenhuis
 
E
Cohen
 
BL
, et al. 
Lunatic Fringe-mediated Notch signaling is required for lung alveogenesis.
Am J Physiol Lung Cell Mol Physiol
2010
, vol. 
298
 
1
(pg. 
L45
-
L56
)
36
Tan
 
JB
Xu
 
K
Cretegny
 
K
, et al. 
Lunatic and manic fringe cooperatively enhance marginal zone B cell precursor competition for delta-like 1 in splenic endothelial niches.
Immunity
2009
, vol. 
30
 
2
(pg. 
254
-
263
)
37
Visan
 
I
Tan
 
JB
Yuan
 
JS
Harper
 
JA
Koch
 
U
Guidos
 
CJ
Regulation of T lymphopoiesis by Notch1 and Lunatic fringe-mediated competition for intrathymic niches.
Nat Immunol
2006
, vol. 
7
 
6
(pg. 
634
-
643
)
38
Tsukumo
 
S
Hirose
 
K
Maekawa
 
Y
Kishihara
 
K
Yasutomo
 
K
Lunatic fringe controls T cell differentiation through modulating notch signaling.
J Immunol
2006
, vol. 
177
 
12
(pg. 
8365
-
8371
)
39
Koch
 
U
Lacombe
 
TA
Holland
 
D
, et al. 
Subversion of the T/B lineage decision in the thymus by lunatic fringe-mediated inhibition of Notch-1.
Immunity
2001
, vol. 
15
 
2
(pg. 
225
-
236
)
40
Acar
 
M
Jafar-Nejad
 
H
Takeuchi
 
H
, et al. 
Rumi is a CAP10 domain glycosyltransferase that modifies Notch and is required for Notch signaling.
Cell
2008
, vol. 
132
 
2
(pg. 
247
-
258
)
41
Kageyama
 
R
Ohtsuka
 
T
Kobayashi
 
T
The Hes gene family: repressors and oscillators that orchestrate embryogenesis.
Development
2007
, vol. 
134
 
7
(pg. 
1243
-
1251
)
42
Wendorff
 
AA
Koch
 
U
Wunderlich
 
FT
, et al. 
Hes1 is a critical but context-dependent mediator of canonical Notch signaling in lymphocyte development and transformation.
Immunity
2010
, vol. 
33
 
5
(pg. 
671
-
684
)
43
Talora
 
C
Campese
 
AF
Bellavia
 
D
, et al. 
Pre-TCR-triggered ERK signalling-dependent downregulation of E2A activity in Notch3-induced T-cell lymphoma.
EMBO Rep
2003
, vol. 
4
 
11
(pg. 
1067
-
1072
)
44
Gonzalez-Garcia
 
S
Garcia-Peydro
 
M
Martin-Gayo
 
E
, et al. 
CSL-MAML-dependent Notch1 signaling controls T lineage-specific IL-7Ralpha gene expression in early human thymopoiesis and leukemia.
J Exp Med
2009
, vol. 
206
 
4
(pg. 
779
-
791
)
45
Bigas
 
A
Robert-Moreno
 
A
Espinosa
 
L
The Notch pathway in the developing hematopoietic system.
Int J Dev Biol
2010
, vol. 
54
 
6
(pg. 
1175
-
1188
)
46
Medvinsky
 
A
Rybtsov
 
S
Taoudi
 
S
Embryonic origin of the adult hematopoietic system: advances and questions.
Development
2011
, vol. 
138
 
6
(pg. 
1017
-
1031
)
47
Ferkowicz
 
MJ
Yoder
 
MC
Blood island formation: longstanding observations and modern interpretations.
Exp Hematol
2005
, vol. 
33
 
9
(pg. 
1041
-
1047
)
48
Ueno
 
H
Weissman
 
IL
Clonal analysis of mouse development reveals a polyclonal origin for yolk sac blood islands.
Dev Cell
2006
, vol. 
11
 
4
(pg. 
519
-
533
)
49
Palis
 
J
Yoder
 
MC
Yolk-sac hematopoiesis: the first blood cells of mouse and man.
Exp Hematol
2001
, vol. 
29
 
8
(pg. 
927
-
936
)
50
Choi
 
K
Kennedy
 
M
Kazarov
 
A
Papadimitriou
 
JC
Keller
 
G
A common precursor for hematopoietic and endothelial cells.
Development
1998
, vol. 
125
 
4
(pg. 
725
-
732
)
51
Fehling
 
HJ
Lacaud
 
G
Kubo
 
A
, et al. 
Tracking mesoderm induction and its specification to the hemangioblast during embryonic stem cell differentiation.
Development
2003
, vol. 
130
 
17
(pg. 
4217
-
4227
)
52
Dieterlen-Lievre
 
F
Beaupain
 
D
Martin
 
C
Origin of erythropoietic stem cells in avian development: shift from the yolk sac to an intraembryonic site.
Ann Immunol (Paris)
1976
, vol. 
127
 
6
(pg. 
857
-
863
)
53
Dieterlen-Lievre
 
F
Pardanaud
 
L
Yassine
 
F
Cormier
 
F
Early haemopoietic stem cells in the avian embryo.
J Cell Sci Suppl
1988
, vol. 
10
 (pg. 
29
-
44
)
54
Cormier
 
F
Dieterlen-Lievre
 
F
The wall of the chick embryo aorta harbours M-CFC, G-CFC, GM-CFC and BFU-E.
Development
1988
, vol. 
102
 
2
(pg. 
279
-
285
)
55
Caprioli
 
A
Minko
 
K
Drevon
 
C
Eichmann
 
A
Dieterlen-Lievre
 
F
Jaffredo
 
T
Hemangioblast commitment in the avian allantois: cellular and molecular aspects.
Dev Biol
2001
, vol. 
238
 
1
(pg. 
64
-
78
)
56
Kanatsu
 
M
Nishikawa
 
SI
In vitro analysis of epiblast tissue potency for hematopoietic cell differentiation.
Development
1996
, vol. 
122
 
3
(pg. 
823
-
830
)
57
Yoder
 
MC
Hiatt
 
K
Mukherjee
 
P
In vivo repopulating hematopoietic stem cells are present in the murine yolk sac at day 9.0 postcoitus.
Proc Natl Acad Sci U S A
1997
, vol. 
94
 
13
(pg. 
6776
-
6780
)
58
Alvarez-Silva
 
M
Belo-Diabangouaya
 
P
Salaun
 
J
Dieterlen-Lievre
 
F
Mouse placenta is a major hematopoietic organ.
Development
2003
, vol. 
130
 
22
(pg. 
5437
-
5444
)
59
Gekas
 
C
Dieterlen-Lievre
 
F
Orkin
 
SH
Mikkola
 
HK
The placenta is a niche for hematopoietic stem cells.
Dev Cell
2005
, vol. 
8
 
3
(pg. 
365
-
375
)
60
Ottersbach
 
K
Dzierzak
 
E
The murine placenta contains hematopoietic stem cells within the vascular labyrinth region.
Dev Cell
2005
, vol. 
8
 
3
(pg. 
377
-
387
)
61
Medvinsky
 
A
Dzierzak
 
E
Definitive hematopoiesis is autonomously initiated by the AGM region.
Cell
1996
, vol. 
86
 (pg. 
897
-
906
)
62
de Bruijn
 
MF
Ma
 
X
Robin
 
C
Ottersbach
 
K
Sanchez
 
MJ
Dzierzak
 
E
Hematopoietic stem cells localize to the endothelial cell layer in the midgestation mouse aorta.
Immunity
2002
, vol. 
16
 
5
(pg. 
673
-
683
)
63
Samokhvalov
 
IM
Samokhvalova
 
NI
Nishikawa
 
S
Cell tracing shows the contribution of the yolk sac to adult haematopoiesis.
Nature
2007
, vol. 
446
 
7139
(pg. 
1056
-
1061
)
64
Chen
 
MJ
Yokomizo
 
T
Zeigler
 
BM
Dzierzak
 
E
Speck
 
NA
Runx1 is required for the endothelial to haematopoietic cell transition but not thereafter.
Nature
2009
, vol. 
457
 
7231
(pg. 
887
-
891
)
65
Zovein
 
AC
Hofmann
 
JJ
Lynch
 
M
, et al. 
Fate tracing reveals the endothelial origin of hematopoietic stem cells.
Cell Stem Cell
2008
, vol. 
3
 
6
(pg. 
625
-
636
)
66
Lawson
 
ND
Scheer
 
N
Pham
 
VN
, et al. 
Notch signaling is required for arterial-venous differentiation during embryonic vascular development.
Development
2001
, vol. 
128
 
19
(pg. 
3675
-
3683
)
67
Duarte
 
A
Hirashima
 
M
Benedito
 
R
, et al. 
Dosage-sensitive requirement for mouse Dll4 in artery development.
Genes Dev
2004
, vol. 
18
 
20
(pg. 
2474
-
2478
)
68
Krebs
 
LT
Shutter
 
JR
Tanigaki
 
K
Honjo
 
T
Stark
 
KL
Gridley
 
T
Haploinsufficient lethality and formation of arteriovenous malformations in Notch pathway mutants.
Genes Dev
2004
, vol. 
18
 
20
(pg. 
2469
-
2473
)
69
Kumano
 
K
Chiba
 
S
Kunisato
 
A
, et al. 
Notch1 but not Notch2 is essential for generating hematopoietic stem cells from endothelial cells.
Immunity
2003
, vol. 
18
 
5
(pg. 
699
-
711
)
70
Robert-Moreno
 
A
Espinosa
 
L
de la Pompa
 
JL
Bigas
 
A
RBPjkappa-dependent Notch function regulates Gata2 and is essential for the formation of intra-embryonic hematopoietic cells.
Development
2005
, vol. 
132
 
5
(pg. 
1117
-
1126
)
71
Burns
 
CE
Traver
 
D
Mayhall
 
E
Shepard
 
JL
Zon
 
LI
Hematopoietic stem cell fate is established by the Notch-Runx pathway.
Genes Dev
2005
, vol. 
19
 
19
(pg. 
2331
-
2342
)
72
You
 
LR
Lin
 
FJ
Lee
 
CT
DeMayo
 
FJ
Tsai
 
MJ
Tsai
 
SY
Suppression of Notch signalling by the COUP-TFII transcription factor regulates vein identity.
Nature
2005
, vol. 
435
 
7038
(pg. 
98
-
104
)
73
Sorensen
 
LK
Brooke
 
BS
Li
 
DY
Urness
 
LD
Loss of distinct arterial and venous boundaries in mice lacking endoglin, a vascular-specific TGFbeta coreceptor.
Dev Biol
2003
, vol. 
261
 
1
(pg. 
235
-
250
)
74
Urness
 
LD
Sorensen
 
LK
Li
 
DY
Arteriovenous malformations in mice lacking activin receptor-like kinase-1.
Nat Genet
2000
, vol. 
26
 
3
(pg. 
328
-
331
)
75
Xue
 
Y
Gao
 
X
Lindsell
 
CE
, et al. 
Embryonic lethality and vascular defects in mice lacking the Notch ligand Jagged1.
Hum Mol Genet
1999
, vol. 
8
 
5
(pg. 
723
-
730
)
76
Robert-Moreno
 
A
Guiu
 
J
Ruiz-Herguido
 
C
, et al. 
Impaired embryonic haematopoiesis yet normal arterial development in the absence of the Notch ligand Jagged1.
EMBO J
2008
, vol. 
27
 
13
(pg. 
1886
-
1895
)
77
Robert-Moreno
 
A
Espinosa
 
L
Sanchez
 
MJ
de la Pompa
 
JL
Bigas
 
A
The notch pathway positively regulates programmed cell death during erythroid differentiation.
Leukemia
2007
, vol. 
21
 
7
(pg. 
1496
-
1503
)
78
Hadland
 
BK
Huppert
 
SS
Kanungo
 
J
, et al. 
A requirement for Notch1 distinguishes 2 phases of definitive hematopoiesis during development.
Blood
2004
, vol. 
104
 
10
(pg. 
3097
-
3105
)
79
Bertrand
 
JY
Cisson
 
JL
Stachura
 
DL
Traver
 
D
Notch signaling distinguishes 2 waves of definitive hematopoiesis in the zebrafish embryo.
Blood
2010
, vol. 
115
 
14
(pg. 
2777
-
2783
)
80
Harman
 
BC
Jenkinson
 
WE
Parnell
 
SM
Rossi
 
SW
Jenkinson
 
EJ
Anderson
 
G
T/B lineage choice occurs prior to intrathymic Notch signaling.
Blood
2005
, vol. 
106
 
3
(pg. 
886
-
892
)
81
Milner
 
LA
Kopan
 
R
Martin
 
DI
Bernstein
 
ID
A human homologue of the Drosophila developmental gene, Notch, is expressed in CD34+ hematopoietic precursors.
Blood
1994
, vol. 
83
 
8
(pg. 
2057
-
2062
)
82
Milner
 
LA
Bigas
 
A
Kopan
 
R
Brashem-Stein
 
C
Bernstein
 
ID
Martin
 
DI
Inhibition of granulocytic differentiation by mNotch1.
Proc Natl Acad Sci U S A
1996
, vol. 
93
 (pg. 
13014
-
13019
)
83
Bigas
 
A
Martin
 
DI
Milner
 
LA
Notch1 and Notch2 inhibit myeloid differentiation in response to different cytokines.
Mol Cell Biol
1998
, vol. 
18
 
4
(pg. 
2324
-
2333
)
84
Calvi
 
LM
Adams
 
GB
Weibrecht
 
KW
, et al. 
Osteoblastic cells regulate the haematopoietic stem cell niche.
Nature
2003
, vol. 
425
 
6960
(pg. 
841
-
846
)
85
Maillard
 
I
Koch
 
U
Dumortier
 
A
, et al. 
Canonical notch signaling is dispensable for the maintenance of adult hematopoietic stem cells.
Cell Stem Cell
2008
, vol. 
2
 
4
(pg. 
356
-
366
)
86
Radtke
 
F
Wilson
 
A
Stark
 
G
, et al. 
Deficient T cell fate specification in mice with an induced inactivation of Notch1.
Immunity
1999
, vol. 
10
 
5
(pg. 
547
-
558
)
87
Varnum-Finney
 
B
Halasz
 
LM
Sun
 
M
Gridley
 
T
Radtke
 
F
Bernstein
 
ID
Notch2 governs the rate of generation of mouse long- and short-term repopulating stem cells.
J Clin Invest
2011
, vol. 
121
 
3
(pg. 
1207
-
1216
)
88
Besseyrias
 
V
Fiorini
 
E
Strobl
 
LJ
, et al. 
Hierarchy of Notch-Delta interactions promoting T cell lineage commitment and maturation.
J Exp Med
2007
, vol. 
204
 
2
(pg. 
331
-
343
)
89
Heng
 
TS
Painter
 
MW
The Immunological Genome Project: networks of gene expression in immune cells.
Nat Immunol
2008
, vol. 
9
 
10
(pg. 
1091
-
1094
)
90
Pui
 
JC
Allman
 
D
Xu
 
L
, et al. 
Notch1 expression in early lymphopoiesis influences B versus T lineage determination.
Immunity
1999
, vol. 
11
 
3
(pg. 
299
-
308
)
91
Aifantis
 
I
Raetz
 
E
Buonamici
 
S
Molecular pathogenesis of T-cell leukaemia and lymphoma.
Nat Rev Immunol
2008
, vol. 
8
 
5
(pg. 
380
-
390
)
92
Aster
 
JC
Deregulated NOTCH signaling in acute T-cell lymphoblastic leukemia/lymphoma: new insights, questions, and opportunities.
Int J Hematol
2005
, vol. 
82
 
4
(pg. 
295
-
301
)
93
Puente
 
XS
Pinyol
 
M
Quesada
 
V
, et al. 
Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia.
Nature
2011
, vol. 
475
 
7354
(pg. 
101
-
105
)
94
Tanigaki
 
K
Han
 
H
Yamamoto
 
N
, et al. 
Notch-RBP-J signaling is involved in cell fate determination of marginal zone B cells.
Nat Immunol
2002
, vol. 
3
 
5
(pg. 
443
-
450
)
95
Saito
 
T
Chiba
 
S
Ichikawa
 
M
, et al. 
Notch2 is preferentially expressed in mature B cells and indispensable for marginal zone B lineage development.
Immunity
2003
, vol. 
18
 
5
(pg. 
675
-
685
)
96
Dallas
 
MH
Varnum-Finney
 
B
Delaney
 
C
Kato
 
K
Bernstein
 
ID
Density of the Notch ligand Delta1 determines generation of B and T cell precursors from hematopoietic stem cells.
J Exp Med
2005
, vol. 
201
 
9
(pg. 
1361
-
1366
)
97
Kim
 
YW
Koo
 
BK
Jeong
 
HW
, et al. 
Defective Notch activation in microenvironment leads to myeloproliferative disease.
Blood
2008
, vol. 
112
 
12
(pg. 
4628
-
4638
)
98
Klinakis
 
A
Lobry
 
C
Abdel-Wahab
 
O
, et al. 
A novel tumour-suppressor function for the Notch pathway in myeloid leukaemia.
Nature
2011
, vol. 
473
 
7346
(pg. 
230
-
233
)
99
Qyang
 
Y
Chambers
 
SM
Wang
 
P
, et al. 
Myeloproliferative disease in mice with reduced presenilin gene dosage: effect of gamma-secretase blockage.
Biochemistry
2004
, vol. 
43
 
18
(pg. 
5352
-
5359
)
100
Yao
 
D
Huang
 
Y
Huang
 
X
, et al. 
Protein O-fucosyltransferase 1 (Pofut1) regulates lymphoid and myeloid homeostasis through modulation of Notch receptor ligand interactions.
Blood
2011
, vol. 
117
 
21
(pg. 
5652
-
5662
)
101
Mercher
 
T
Cornejo
 
MG
Sears
 
C
, et al. 
Notch signaling specifies megakaryocyte development from hematopoietic stem cells.
Cell Stem Cell
2008
, vol. 
3
 
3
(pg. 
314
-
326
)
102
Demehri
 
S
Liu
 
Z
Lee
 
J
, et al. 
Notch-deficient skin induces a lethal systemic B-lymphoproliferative disorder by secreting TSLP, a sentinel for epidermal integrity.
PLoS Biol
2008
, vol. 
6
 
5
pg. 
e123
 
103
Dumortier
 
A
Durham
 
AD
Di Piazza
 
M
, et al. 
Atopic dermatitis-like disease and associated lethal myeloproliferative disorder arise from loss of Notch signaling in the murine skin.
PLoS One
2010
, vol. 
5
 
2
pg. 
e9258
 
104
Mansson
 
R
Hultquist
 
A
Luc
 
S
, et al. 
Molecular evidence for hierarchical transcriptional lineage priming in fetal and adult stem cells and multipotent progenitors.
Immunity
2007
, vol. 
26
 
4
(pg. 
407
-
419
)
105
Katsura
 
Y
Kawamoto
 
H
Stepwise lineage restriction of progenitors in lympho-myelopoiesis.
Int Rev Immunol
2001
, vol. 
20
 
1
(pg. 
1
-
20
)
106
Kawamoto
 
H
Katsura
 
Y
A new paradigm for hematopoietic cell lineages: revision of the classical concept of the myeloid-lymphoid dichotomy.
Trends Immunol
2009
, vol. 
30
 
5
(pg. 
193
-
200
)
107
Lai
 
AY
Kondo
 
M
Asymmetrical lymphoid and myeloid lineage commitment in multipotent hematopoietic progenitors.
J Exp Med
2006
, vol. 
203
 
8
(pg. 
1867
-
1873
)
108
Varnum-Finney
 
B
Xu
 
L
Brashem-Stein
 
C
, et al. 
Pluripotent, cytokine-dependent, hematopoietic stem cells are immortalized by constitutive notch1 signaling.
Nat Med
2000
, vol. 
6
 
11
(pg. 
1278
-
1281
)
109
Ohishi
 
K
Varnum-Finney
 
B
Bernstein
 
ID
Delta-1 enhances marrow and thymus repopulating ability of human CD34(+)CD38(−) cord blood cells.
J Clin Invest
2002
, vol. 
110
 
8
(pg. 
1165
-
1174
)
110
Delaney
 
C
Heimfeld
 
S
Brashem-Stein
 
C
Voorhies
 
H
Manger
 
RL
Bernstein
 
ID
Notch-mediated expansion of human cord blood progenitor cells capable of rapid myeloid reconstitution.
Nat Med
2010
, vol. 
16
 
2
(pg. 
232
-
236
)
111
Butler
 
JM
Nolan
 
DJ
Vertes
 
EL
, et al. 
Endothelial cells are essential for the self-renewal and repopulation of Notch-dependent hematopoietic stem cells.
Cell Stem Cell
2010
, vol. 
6
 
3
(pg. 
251
-
264
)
112
Karanu
 
FN
Murdoch
 
B
Gallacher
 
L
, et al. 
The notch ligand jagged-1 represents a novel growth factor of human hematopoietic stem cells.
J Exp Med
2000
, vol. 
192
 
9
(pg. 
1365
-
1372
)
113
Carlesso
 
N
Aster
 
JC
Sklar
 
J
Scadden
 
DT
Notch1-induced delay of human hematopoietic progenitor cell differentiation is associated with altered cell cycle kinetics.
Blood
1999
, vol. 
93
 
3
(pg. 
838
-
848
)
114
Shimojo
 
H
Ohtsuka
 
T
Kageyama
 
R
Dynamic expression of notch signaling genes in neural stem/progenitor cells.
Front Neurosci
2011
, vol. 
5
 pg. 
78
 
115
Yoshiura
 
S
Ohtsuka
 
T
Takenaka
 
Y
Nagahara
 
H
Yoshikawa
 
K
Kageyama
 
R
Ultradian oscillations of Stat, Smad, and Hes1 expression in response to serum.
Proc Natl Acad Sci U S A
2007
, vol. 
104
 
27
(pg. 
11292
-
11297
)
116
Shimojo
 
H
Ohtsuka
 
T
Kageyama
 
R
Oscillations in notch signaling regulate maintenance of neural progenitors.
Neuron
2008
, vol. 
58
 
1
(pg. 
52
-
64
)
117
Kageyama
 
R
Ohtsuka
 
T
Shimojo
 
H
Imayoshi
 
I
Dynamic Notch signaling in neural progenitor cells and a revised view of lateral inhibition.
Nat Neurosci
2008
, vol. 
11
 
11
(pg. 
1247
-
1251
)
118
Bertrand
 
N
Castro
 
DS
Guillemot
 
F
Proneural genes and the specification of neural cell types.
Nat Rev Neurosci
2002
, vol. 
3
 
7
(pg. 
517
-
530
)
119
Kopan
 
R
Ilagan
 
MX
The canonical Notch signaling pathway: unfolding the activation mechanism.
Cell
2009
, vol. 
137
 
2
(pg. 
216
-
233
)
120
Yamamoto
 
N
Tanigaki
 
K
Han
 
H
Hiai
 
H
Honjo
 
T
Notch/RBP-J signaling regulates epidermis/hair fate determination of hair follicular stem cells.
Curr Biol
2003
, vol. 
13
 
4
(pg. 
333
-
338
)
121
Demehri
 
S
Kopan
 
R
Notch signaling in bulge stem cells is not required for selection of hair follicle fate.
Development
2009
, vol. 
136
 
6
(pg. 
891
-
896
)
122
Estrach
 
S
Ambler
 
CA
Lo Celso
 
C
Hozumi
 
K
Watt
 
FM
Jagged 1 is a beta-catenin target gene required for ectopic hair follicle formation in adult epidermis.
Development
2006
, vol. 
133
 
22
(pg. 
4427
-
4438
)
123
Rangarajan
 
A
Talora
 
C
Okuyama
 
R
, et al. 
Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation.
EMBO J
2001
, vol. 
20
 
13
(pg. 
3427
-
3436
)
124
Nicolas
 
M
Wolfer
 
A
Raj
 
K
, et al. 
Notch1 functions as a tumor suppressor in mouse skin.
Nat Genet
2003
, vol. 
33
 
3
(pg. 
416
-
421
)
125
Radtke
 
F
Raj
 
K
The role of Notch in tumorigenesis: oncogene or tumour suppressor?
Nat Rev Cancer
2003
, vol. 
3
 
10
(pg. 
756
-
767
)
126
Blanpain
 
C
Lowry
 
WE
Pasolli
 
HA
Fuchs
 
E
Canonical notch signaling functions as a commitment switch in the epidermal lineage.
Genes Dev
2006
, vol. 
20
 
21
(pg. 
3022
-
3035
)
127
Demehri
 
S
Turkoz
 
A
Kopan
 
R
Epidermal Notch1 loss promotes skin tumorigenesis by impacting the stromal microenvironment.
Cancer Cell
2009
, vol. 
16
 
1
(pg. 
55
-
66
)
128
Pan
 
Y
Lin
 
MH
Tian
 
X
, 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
, vol. 
7
 
5
(pg. 
731
-
743
)
129
Weber
 
S
Niessen
 
MT
Prox
 
J
, et al. 
The disintegrin/metalloproteinase Adam10 is essential for epidermal integrity and Notch-mediated signaling.
Development
2011
, vol. 
138
 
3
(pg. 
495
-
505
)
130
Sato
 
T
van Es
 
JH
Snippert
 
HJ
, et al. 
Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts.
Nature
2011
, vol. 
469
 
7330
(pg. 
415
-
418
)
131
Tian
 
H
Biehs
 
B
Warming
 
S
, et al. 
A reserve stem cell population in small intestine renders Lgr5-positive cells dispensable.
Nature
2011
, vol. 
478
 
7368
(pg. 
255
-
259
)
132
Li
 
L
Clevers
 
H
Coexistence of quiescent and active adult stem cells in mammals.
Science
2010
, vol. 
327
 
5965
(pg. 
542
-
545
)
133
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
, vol. 
435
 
7044
(pg. 
964
-
968
)
134
Jensen
 
J
Pedersen
 
EE
Galante
 
P
, et al. 
Control of endodermal endocrine development by Hes-1.
Nat Genet
2000
, vol. 
24
 
1
(pg. 
36
-
44
)
135
Fre
 
S
Hannezo
 
E
Sale
 
S
, et al. 
Notch lineages and activity in intestinal stem cells determined by a new set of knock-in mice.
PLoS One
2011
, vol. 
6
 
10
pg. 
e25785
 
136
Pellegrinet
 
L
Rodilla
 
V
Liu
 
Z
, et al. 
Dll1- and dll4-mediated notch signaling are required for homeostasis of intestinal stem cells.
Gastroenterology
2011
, vol. 
140
 
4
(pg. 
1230
-
1237
)
137
Mathur
 
D
Bost
 
A
Driver
 
I
Ohlstein
 
B
A transient niche regulates the specification of Drosophila intestinal stem cells.
Science
2010
, vol. 
327
 
5962
(pg. 
210
-
213
)
138
Clements
 
WK
Kim
 
AD
Ong
 
KG
Moore
 
JC
Lawson
 
ND
Traver
 
D
A somitic Wnt16/Notch pathway specifies haematopoietic stem cells.
Nature
2011
, vol. 
474
 
7350
(pg. 
220
-
224
)
139
Carlson
 
ME
Hsu
 
M
Conboy
 
IM
Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells.
Nature
2008
, vol. 
454
 
7203
(pg. 
528
-
532
)
140
Conboy
 
IM
Rando
 
TA
Aging, stem cells and tissue regeneration: lessons from muscle.
Cell Cycle
2005
, vol. 
4
 
3
(pg. 
407
-
410
)
141
Kageyama
 
R
Niwa
 
Y
Shimojo
 
H
Kobayashi
 
T
Ohtsuka
 
T
Ultradian oscillations in Notch signaling regulate dynamic biological events.
Curr Top Dev Biol
2010
, vol. 
92
 (pg. 
311
-
331
)
Sign in via your Institution