Regulating the Notch pathway in embryonic, adult and old stem cells

https://doi.org/10.1016/j.coph.2007.02.004Get rights and content

The Notch pathway represents a highly conserved signaling network, which is critical to both embryonic skeletal muscle formation and regeneration in the adult. In addition to skeletal muscle, Notch also regulates the formation and maintenance of various organ systems, such as brain, blood and intestine, in evolutionary distinct vertebrate and invertebrate species. The Notch network ‘cross talks’ with all other key cell-fate determinants, such as the Wnt (Wingless), TGF-β/BMP, Hh and RTK/Ras pathways. Hence, modulating the intensity of Notch resonates through multiple regulatory circuitries, and exerts profound effects on cell behaviour. Therefore, various approaches to the targeted manipulation of Notch have been developed (e.g. genetic constructs, antibodies, RNA interference, receptor decoys and γ-secretase inhibitors). These tools might be used to broaden our understanding of this pathway in regulating responses of embryonic and adult stem cell subsets, and to develop therapeutic approaches against Notch-based diseases (e.g. Alzheimer's, Alagille Syndrome, various cancers and other disease states).

Introduction

Since its discovery approximately 90 years ago, the Notch pathway (from ‘notched’ wingblade phenotype in fruit flies defective for this locus) has been extensively studied and exploited within arenas of developmental biology, regenerative medicine and oncology. Notch is considered to be almost universally conserved in its role of controlling embryonic organogenesis and postnatal tissue repair [1, 2, 3•]. The variable strength of Notch signaling represents a highly reliable molecular mechanism that regulates cell-fate determination, and enables formation of tissues and organs from initially equivalent groups of stem cells. Likewise, we know that Notch signaling is involved with embryonic, postnatal, aged and oncogenic properties of stem and progenitor cells.

The interactions between Notch signaling and other key molecular networks (e.g. transforming growth factor/bone morphogenetic protein-4 and Wingless [Wnt]) have also been conserved in postnatal regeneration of multiple tissues, and are implicated in Notch-imposed control of stem cell responses [4, 5, 6]. Consequentially, our ability to regulate Notch signalling, both positively and negatively, has wide implications for many areas of experimental biology, ranging from the bourgeoning field of embryonic stem cell research to the studies of aging. Here, we review the role of Notch signaling, from embryonic to specific adult stem cell subsets. We also outline recent advances in our ability to manipulate the Notch pathway, and discuss both their clinical and research implications.

Section snippets

Notch signaling

To date, four isoforms of the Notch receptor have been identified in mammals (Notch1–4), whereas two isoforms are reported in Caenorhabditis elegans (LIN-12 and GLP-1) and one in Drosophila melanogaster (Notch) [7, 8•]. As a precursor protein, immature Notch undergoes furin-like proteolytic processing (S1 cleavage) in the trans-golgi, before being presented as a single-pass transmembrane receptor on the cell surface [9]. The mature Notch receptor is a heterodimer, consisting of a large

Stem cells and their niche in young and old organisms: role of the Notch pathway

A diversity of stem cell subsets require Notch signaling for the productive maintenance of tissues [11]. However, with advancing age, stem-cell-regulating extrinsic cues become adversely altered, both systemically and in the local organ environment. This phenomenon precludes productive tissue repair, and has been demonstrated in heterochronic tissue transplantation and parabiosis studies, as well as with embryonic stem cells [33, 34, 35]. The age-associated decline in stem cell regeneration

Therapeutic potential of embryonic stem cells

ESCs possess the distinct and defining features of both unlimited self-renewal and pluripotency [45, 46]. These characteristics endow ESCs with tremendous neo-organogenesis potential and have consequently generated great interest in ESC-based cell-replacement therapies. Understanding the molecular events that govern self-renewal and tissue-specific differentiation of ESCs is critical to controlling their productive regenerative properties. In vitro multilineage differentiation of ESCs is

The Notch pathway and the directed differentiation of embryonic stem cells

The function of endogenous Notch signaling in mammalian ESCs is just beginning to be understood in detail. mRNAs encoding Notch1–3 and DLL1 are present at high levels in hESCs [54, 55]; additionally, both Notch3 and active-form γ-secretase components are reported to be highly expressed in mESCs and hESCs [56, 57]. Conversely, Notch2 is expressed at much lower levels [54]. Notch signaling is thought to be relatively inactive in undifferentiated hESCs, and not necessary for their propagation.

Pharmacological manipulation of the Notch pathway

Notch signaling can be inhibited through various means, including monoclonal antibodies, RNA interference, antisense Notch, receptor and mastermind-like 1 (MAML1) decoys, and the use of γ-secretase inhibitors (GSIs) [69]. Experimental manipulation of the strength of Notch signaling has provided important data on somitogenesis, myogenesis, neurogenesis and the transdifferentiation of bone marrow mesenchymal stem cells into muscle cells [4, 11, 70••, 71, 72]. In particular, GSIs have become an

Conclusions

After many decades of productive research, the Notch pathway remains at the forefront of modern biomedical science and continues to inform us of the mechanisms controlling cell behavior. The conservation of this signaling network between evolutionarily distinct species and different cell types is truly remarkable, and productive Notch activity is critically important not only for the formation, but also for the maintenance and repair, of numerous organs and tissues. Alterations in Notch

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

The authors would like to acknowledge, and apologise to, the numerous authors who have made important contributions to the fields addressed in this review, but whose work was not included due to the limited scope and space restrictions of this article.

References (86)

  • E. Andersson et al.

    Identification of intrinsic determinants of midbrain dopamine neurons

    Cell

    (2006)
  • J.Y. Rho et al.

    Transcriptional profiling of the developmentally important signalling pathways in human embryonic stem cells

    Hum Reprod

    (2006)
  • M. Nemir et al.

    Induction of cardiogenesis in embryonic stem cells via downregulation of Notch1 signaling

    Circ Res

    (2006)
  • T. Schroeder et al.

    Activated Notch1 alters differentiation of embryonic stem cells into mesodermal cell lineages at multiple stages of development

    Mech Dev

    (2006)
  • N.R. Treff et al.

    Differentiation of embryonic stem cells conditionally expressing neurogenin 3

    Stem Cells

    (2006)
  • L. Miele et al.

    Notch signaling in cancer

    Curr Mol Med

    (2006)
  • M. Dezawa et al.

    Bone marrow stromal cells generate muscle cells and repair muscle degeneration

    Science

    (2005)
  • J.Z. Qin et al.

    p53-independent NOXA induction overcomes apoptotic resistance of malignant melanomas

    Mol Cancer Ther

    (2004)
  • C. Cornbrooks et al.

    Delta expression in post-mitotic neurons identifies distinct subsets of adult-specific lineages in Drosophila

    J Neurobiol

    (2006)
  • J. Krishnamurthy et al.

    p16INK4a induces an age-dependent decline in islet regenerative potential

    Nature

    (2006)
  • S. Artavanis-Tsakonas et al.

    Notch signaling: cell fate control and signal integration in development

    Science

    (1999)
  • S.J. Bray

    Notch signalling: a simple pathway becomes complex

    Nat Rev Mol Cell Biol

    (2006)
  • A.V. Molofsky et al.

    Diverse mechanisms regulate stem cell self-renewal

    Curr Opin Cell Biol

    (2004)
  • S. Kamakura et al.

    Hes binding to STAT3 mediates crosstalk between Notch and JAK-STAT signalling

    Nat Cell Biol

    (2004)
  • F. Schweisguth

    Regulation of notch signaling activity

    Curr Biol

    (2004)
  • C.M. Blaumueller et al.

    Intracellular cleavage of Notch leads to a heterodimeric receptor on the plasma membrane

    Cell

    (1997)
  • M.D. Rand et al.

    Calcium depletion dissociates and activates heterodimeric notch receptors

    Mol Cell Biol

    (2000)
  • E. Ladi et al.

    The divergent DSL ligand Dll3 does not activate Notch signaling but cell autonomously attenuates signaling induced by other DSL ligands

    J Cell Biol

    (2005)
  • S. Shi et al.

    Protein O-fucosyltransferase 1 is an essential component of Notch signaling pathways

    Proc Natl Acad Sci USA

    (2003)
  • T. Okajima et al.

    Modulation of notch-ligand binding by protein O-fucosyltransferase 1 and fringe

    J Biol Chem

    (2003)
  • N. Haines et al.

    Glycosylation regulates Notch signalling

    Nat Rev Mol Cell Biol

    (2003)
  • C. Brou et al.

    A novel proteolytic cleavage involved in Notch signaling: the role of the disintegrin-metalloprotease TACE

    Mol Cell

    (2000)
  • F. Radtke et al.

    Notch regulation of lymphocyte development and function

    Nat Immunol

    (2004)
  • M.E. Fortini

    Gamma-secretase-mediated proteolysis in cell-surface-receptor signalling

    Nat Rev Mol Cell Biol

    (2002)
  • H.Y. Kao et al.

    A histone deacetylase corepressor complex regulates the Notch signal transduction pathway

    Genes Dev

    (1998)
  • E.C. Lai

    Keeping a good pathway down: transcriptional repression of Notch pathway target genes by CSL proteins

    EMBO Rep

    (2002)
  • M.A. McGill et al.

    Mammalian numb proteins promote Notch1 receptor ubiquitination and degradation of the Notch1 intracellular domain

    J Biol Chem

    (2003)
  • D. Berdnik et al.

    The endocytic protein alpha-Adaptin is required for numb-mediated asymmetric cell division in Drosophila

    Dev Cell

    (2002)
  • T.G. Doyle et al.

    SEL-8, a nuclear protein required for LIN-12 and GLP-1 signaling in Caenorhabditis elegans

    Proc Natl Acad Sci USA

    (2000)
  • A.E. Wallberg et al.

    p300 and PCAF act cooperatively to mediate transcriptional activation from chromatin templates by notch intracellular domains in vitro

    Mol Cell Biol

    (2002)
  • L. Wu et al.

    MAML1, a human homologue of Drosophila mastermind, is a transcriptional co-activator for NOTCH receptors

    Nat Genet

    (2000)
  • T. Iso et al.

    HES and HERP families: multiple effectors of the Notch signaling pathway

    J Cell Physiol

    (2003)
  • F. Radtke et al.

    The role of Notch in tumorigenesis: oncogene or tumour suppressor?

    Nat Rev Cancer

    (2003)
  • Cited by (58)

    • Aging, metabolism and stem cells: Spotlight on muscle stem cells

      2017, Molecular and Cellular Endocrinology
      Citation Excerpt :

      Also, elevated Wnt levels in the circulation of aged individuals promote SC differentiation and reduce the capacity of SCs to self-renew (Brack et al., 2007; Carlson et al., 2009; Brack et al., 2008). These pro-aging effects can be reproduced in young SCs by inhibition of Notch signaling, whereas muscle regeneration in aged mice can be partially rescued by delivery of active Notch (Carlson et al., 2007; Carlson and Conboy, 2007). Some of the most compelling evidences for the impact of age-associated changes in the stem cell niche and/or environment have come from elegant experiments that have experimentally restored “youthful” characteristics to the stem cell environment and thus improved regeneration in aged rodents (Sousa-Victor et al., 2015).

    • 6.13 Tissue engineering of muscle tissue

      2017, Comprehensive Biomaterials II
    • Amygdala-Dependent Fear Memory Consolidation via miR-34a and Notch Signaling

      2014, Neuron
      Citation Excerpt :

      Second, using a targeted bioinformatics approach, complemented with loss- and gain-of-function studies, we establish a link for miRNA-mediated regulation of the Notch pathway as being involved in this consolidation. With Notch pathway function being traditionally viewed through the lens of stem cell differentiation during development (Carlson and Conboy, 2007), this second contribution indicates that signaling pathways normatively used in development are also parsimoniously utilized for adult behavior. Finally, Notch signaling is now being targeted in the treatment of various cancers (Rizzo et al., 2008; Sail and Hadden, 2012), with numerous new therapeutic tools available.

    • Immunohistochemical expression of aberrant Notch-1 signaling in vitiligo: An implication for pathogenesis

      2014, Annals of Diagnostic Pathology
      Citation Excerpt :

      Upon ligand binding, 2 sequential proteolytic events occur to liberate active Notch intracellular domain (NICD), which is then translocated to the nucleus where it binds to the transcriptional repressor, CSL (RBP-J), to activate target genes expression [5]. Notch signaling pathway is an essential cell-cell interaction mechanism that regulates processes such as cell proliferation, cell fate decisions, differentiation, or stem cell maintenance [6]. Notch appears to act as an oncogene in melanocytes, and activation of its signaling may occur during the formation of melanocytic tumors [7].

    View all citing articles on Scopus
    View full text