Elsevier

Bone

Volume 46, Issue 2, February 2010, Pages 281-285
Bone

Review
Notch signaling and the bone marrow hematopoietic stem cell niche

https://doi.org/10.1016/j.bone.2009.08.007Get rights and content

Abstract

Recently there has been increased interest in the regulatory interactions between osteoblasts and cells in the surrounding bone marrow microenvironment. The proximity of hematopoietic stem cells (HSCs) with osteoblastic cells first suggested regulatory interactions, and recent data have highlighted the role of osteoblastic cells in providing a HSC niche. Reports have indicated that direct contact is necessary to mediate the osteoblastic effects and that these effects could be mediated through Notch activation. Notch signaling is important throughout development and also appears to play a critical role in cellular maturation and differentiation of osteoblastic cells and hematopoietic cells as disregulation can lead to bone loss and leukemias, respectively. In this review we discuss the current understanding of Notch signaling and how it functions in hematopoiesis, osteoblastic cells, and the interactions between HSC and their osteoblastic niche.

Introduction

In the late 1970s Schofield used the term niche to define custom microenvironments composed of subset of cells and extracellular substrates required to support localization, survival, and self-renewal of stem cells in vivo [1]. Conceptually the niche can be thought of as a region that can harbor stem cells and maintain balance between self renewal and differentiation. Over the years, a body of evidence has accumulated in a number of animal models that supports the concept of microenvironmental regulation of stem cells [2]. Notch signaling is in many ways ideal for instructive communication between the niche and stem cells as it is highly conserved, requires direct contact of adjacent cells, and Notch signaling can influence cell fate decisions [3]. While HSCs are fairly well understood, only recently data have indicated cellular and molecular components of the bone marrow HSC niche [4], with some of the first evidence suggesting that osteoblastic cells are supportive of HSCs coming from in vitro work [5]. The important role of Notch signaling in hematopoiesis was highlighted by its association with hematological malignancies [6], [7], which initiated a great deal of interest in the role of Notch signaling in HSC regulation and in hematopoietic lineage allocation and differentiation, as we will review. Most recently, in vivo evidence has strongly implicated Notch signaling in regulation of osteoblastic differentiation [8], [9]. Together, these lines of investigation would suggest that Notch signaling in the bone marrow microenvironment could be essential in HSC-osteoblastic regulatory interactions, and in fact, data defining the role of osteoblastic cells in HSC regulation point to Notch signaling as a potential intermediary [10]. In this review we will discuss the Notch signaling pathway and how it functions in the hematopoietic system, osteoblastic cells, and the interaction between HSC and their microenvironment which have emerged so far.

Section snippets

Notch signaling

The canonical Notch Signaling pathway is highly conserved and plays roles in many cellular functions including cellular growth, differentiation, and fate choices [3]. In mammals four Notch receptors (Notch 1–4) and five ligands (Jagged1–2 and Delta-like 1, 3, and 4) have been identified, all of which are single span transmembrane proteins that require cell–cell contact for activation [11]. When a Notch ligand comes in contact with Notch, the external portion of Notch is cleaved by tumor

Notch, leukemias and hematopoietic lineage allocation

Strong evidence for the role of Notch signaling in hematopoiesis is provided by the finding that aberrations in Notch can lead to leukemias. Notch was first linked to T-cell neoplasias in the late 1980s and early 1990s with the identification of the t(7;9) chromosomal translocation which was cloned from a subset of T-cell acute lymphoblastic leukemias (T-ALL) [6], [7]. This translocation resulted in a truncated form of Notch1 termed translocation associated Notch homolog (TAN1), which

In vitro studies

Many independent studies have established that Notch1 and the Notch ligands Jagged1 and Delta-like1 are expressed in osteoblasts [10], [31], [32], [33], [34], [35], [36]. In vitro studies attempting to elucidate the role of Notch signaling in bone have led to contradictory conclusions that Notch activation either impairs [32], [33], [37], [38], [39] or facilitates [34], [40], [41] osteoblastic differentiation. The disparity in these studies could be due to different cellular models, different

Notch and stem cell niches

As a system providing rapid environmental cell-initiated signals to stem cells, the Notch signaling pathway would be predicted to play an important role in stem cell niches. In fact, much data support the role of Notch signaling as a key mediator of niche-stem cell interactions in a number of organisms and in numerous organ systems. For example, Notch is reported to be important in gonadal niches in both worms [46] and flies [47], [48], where Notch signaling may also determine niche size. In

Notch and the microenvironmental control of hematopoiesis

Examples exist of the importance of Notch signaling in relaying instruction from the microenvironment to stem cells in numerous systems, but what about mammalian HSCs? Mammalian HSCs are some of the best characterized stem cells. One of the difficulties in determining the signals involved in HSC regulation by the microenvironment has been that the cellular regulatory components of the bone marrow microenvironment have only recently come to light. Currently the HSC niche is thought to be

Concluding thoughts

Although the verdict is still out on the role of Notch in HSC maintenance, it is evident from in vitro studies that Notch ligands can increase HSC and maintain progenitor populations. It also appears that normal Notch signaling in the bone marrow microenvironment is necessary to maintain normal hematopoiesis. Based on the data available so far, it is unlikely that Notch signaling is the only pathway to regulate HSC within the niche. Many other pathways, such as N-cadherin [50],

Acknowledgments

This work was supported by the National Institutes of Health, National Institute of Diabetes, Digestive and Kidney Diseases (K08 DK064381, R21 DK069563, R01 DK 076876 to L.M.C.), the Wilmot Cancer Scholar Foundation (L.M.C.) and the Pew Foundation (L.M.C.). J.M.W. is supported by T32 AR05345903. The authors thank Matthew J. Hilton, Benjamin J. Frisch and Rebecca L. Porter for their helpful discussions.

References (73)

  • J.M. Weber et al.

    Parathyroid hormone stimulates expression of the Notch ligand Jagged1 in osteoblastic cells

    Bone

    (2006)
  • A. Smerdel-Ramoya et al.

    Connective tissue growth factor enhances osteoblastogenesis in vitro

    J. Biol. Chem.

    (2008)
  • K. Shindo et al.

    Osteogenic differentiation of the mesenchymal progenitor cells, Kusa is suppressed by Notch signaling

    Exp. Cell Res.

    (2003)
  • T. Yamada et al.

    Regulation of osteoclast development by Notch signaling directed to osteoclast precursors and through stromal cells

    Blood

    (2003)
  • S. Bai et al.

    NOTCH1 regulates osteoclastogenesis directly in osteoclast precursors and indirectly via osteoblast lineage cells

    J. Biol. Chem.

    (2008)
  • E.J. Ward et al.

    Stem cells signal to the niche through the Notch pathway in the Drosophila ovary

    Curr. Biol.

    (2006)
  • D. Visnjic et al.

    Hematopoiesis is severely altered in mice with an induced osteoblast deficiency

    Blood

    (2004)
  • A. Colmone et al.

    Beyond angiogenesis: the role of endothelium in the bone marrow vascular niche

    Transl. Res.

    (2008)
  • F.N. Karanu et al.

    Human homologues of Delta-1 and Delta-4 function as mitogenic regulators of primitive human hematopoietic cells

    Blood

    (2001)
  • B. Varnum-Finney et al.

    The Notch ligand, Jagged-1, influences the development of primitive hematopoietic precursor cells

    Blood

    (1998)
  • B. Varnum-Finney et al.

    Combined effects of Notch signaling and cytokines induce a multiple log increase in precursors with lymphoid and myeloid reconstituting ability

    Blood

    (2003)
  • N. Carlesso et al.

    Notch1-induced delay of human hematopoietic progenitor cell differentiation is associated with altered cell cycle kinetics

    Blood

    (1999)
  • L. Li et al.

    The human homolog of rat Jagged1 expressed by marrow stroma inhibits differentiation of 32D cells through interaction with Notch1

    Immunity

    (1998)
  • B. Sacchetti et al.

    Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment

    Cell

    (2007)
  • L. Qin et al.

    Gene expression profiles and transcription factors involved in parathyroid hormone signaling in osteoblasts revealed by microarray and bioinformatics

    J. Biol. Chem.

    (2003)
  • S.J. Mancini et al.

    Jagged1-dependent Notch signaling is dispensable for hematopoietic stem cell self-renewal and differentiation

    Blood

    (2005)
  • I. Maillard et al.

    Canonical notch signaling is dispensable for the maintenance of adult hematopoietic stem cells

    Cell Stem Cell

    (2008)
  • Y.W. Kim et al.

    Defective Notch activation in microenvironment leads to myeloproliferative disease

    Blood

    (2008)
  • F. Arai et al.

    Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche

    Cell

    (2004)
  • S.K. Nilsson et al.

    Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells

    Blood

    (2005)
  • Y. Jung et al.

    Annexin II expressed by osteoblasts and endothelial cells regulates stem cell adhesion, homing, and engraftment following transplantation

    Blood

    (2007)
  • R. Schofield

    The relationship between the spleen colony-forming cell and the haemopoietic stem cell

    Blood Cells

    (1978)
  • A. Spradling et al.

    Stem cells find their niche

    Nature

    (2001)
  • S. Artavanis-Tsakonas et al.

    Notch signaling: cell fate control and signal integration in development

    Science

    (1999)
  • R.S. Taichman et al.

    The role of osteoblasts in the hematopoietic microenvironment

    Stem Cells

    (1998)
  • F. Engin et al.

    Dimorphic effects of Notch signaling in bone homeostasis

    Nat. Med.

    (2008)
  • Cited by (0)

    View full text