Elsevier

Experimental Cell Research

Volume 295, Issue 1, 15 April 2004, Pages 128-137
Experimental Cell Research

CCAAT/enhancer-binding protein-β has a role in osteoblast proliferation and differentiation

https://doi.org/10.1016/j.yexcr.2004.01.004Get rights and content

Abstract

CCAAT/enhancer binding protein β (C/EBPβ) is known to play an important role in the expression of several genes necessary for bone development and homeostasis including osteocalcin, IGF-1, and IL-6. In this study, we show that C/EBPβ protein levels and, consequently, DNA-binding activity are temporally regulated, dramatically decreasing upon differentiation of MC3T3-E1 mouse osteoblasts. Corresponding with these results, the constitutive expression of C/EBPβ LAP in MC3T3-E1 osteoblasts increased proliferation and suppressed osteogenic differentiation. Thus, C/EBPβ LAP not only appears to participate in the regulation of genes associated with mature bone physiology, but is also a critical regulator of osteoblast growth and differentiation.

Introduction

CCAAT/enhancer binding protein (C/EBP) α, β, γ, δ, ε, and ζ comprise a family of basic region-leucine zipper (bZIP) transcription factors (reviewed in Ref. [1]). These proteins dimerize through their leucine zipper domains and bind to DNA through their adjacent basic regions. C/EBPα, β, δ, and ε can activate in vivo transcription from promoters that contain a consensus binding site: 5′-T(T/G)NNGNAA(T/G)-3′ [2]. The in vitro functions of these family members are nearly identical to those of prototypical C/EBP, but the variety of C/EBP family members and their potential for heterodimer formation could provide a large repertoire of transcription factors with complex in vivo regulatory features.

C/EBPβ has been most extensively characterized as an effector in the induction of acute phase and inflammatory genes responsive to LPS, IL-1, or IL-6 ([2], [3], [4], [5], reviewed in Ref. [6]). Consistent with this role, the DNA-binding activity of C/EBPβ is increased by a post-transcriptional mechanism when cells are exposed to LPS, IL-1, or IL-6 [2], [5] and C/EBPβ expression is capable of enhancing transcription from promoters containing elements responsive to those factors [2], [3], [4], [5]. Although the primary mechanism of C/EBPβ regulation within inflammatory responses appears to be post-transcriptional [7], C/EBPβ mRNA levels are also induced by LPS, IL-1, or IL-6 [2]. Thus, there is a circular aspect to C/EBPβ regulation with regulated gene products (e.g. IL-1 and IL-6) regulating expression of the regulator (i.e. C/EBPβ).

Among the post-transcriptional mechanisms for C/EBPβ regulation, covalent modification has been shown to be an important mechanism for modulating its activity. Site-specific phosphorylation through the MAP kinase pathway concomitant with increased C/EBPβ transactivation is stimulated by the ras oncogene [8], as well as stimulation by interferon γ [9] and growth hormone [10]. C/EBPβ activity is also stimulated by site-specific phosphorylation in response to elevated intracellular levels of calcium through a calcium/calmodulin-dependent kinase [11]. TPA, acting through the protein kinase C pathway, also increases C/EBPβ site-specific phosphorylation and activity [12]. This phosphorylation is critical for TGFα-induced hepatocyte proliferation [13]. In vitro studies have identified several protein kinase A and C phosphorylation sites that modulate DNA binding, including one that attenuates DNA binding [14], as does site-specific glycogen synthase kinase 3 phosphorylation [15]. All of these instances of modulation of C/EBPβ activity and phosphorylation involve several phosphorylation sites and protein kinases. Clearly, C/EBPβ can be regulated by a variety of signal transduction pathways, perhaps depending upon the cell type in which it is expressed. More recently, C/EBPβ activity has been reported to be increased by sumoylation [16] and to be attenuated by acetylation [17].

Another level of post-transcriptional regulation for C/EBPβ is provided by the expression of three C/EBPβ isoforms, LAP* (38 kDa), LAP (35 kDa), and LIP (20 kDa), through the use of alternative translational start sites in the C/EBPβ mRNA [18] or proteolysis [19], [20]. The regulation of alternative translational initiation depends upon an evolutionarily conserved upstream open reading frame and the regulation of translation factor eIF activity [21], as well as the CUG-repeat binding protein interactions with the 5′ region of the C/EBPβ mRNA [22]. LAP* and LAP both retain three transcriptional activation domain modules, whereas LIP lacks these activation domain modules [23]. LIP is unable to activate transcription from a C/EBP-dependent promoter-reporter and can inhibit C/EBPβ-mediated transcriptional activation [18]. On the other hand, LIP can activate transcription from the IL-6 promoter with LPS stimulation [24]. Overexpression of LIP has been observed in fetal liver cells [18] and in mammary tumors [25] where it is proposed to inhibit the expression of C/EBP-dependent genes that promote differentiation and inhibit proliferation, thus promoting proliferation and/or a transformed phenotype. LAP* differs from LAP by a short N-terminal extension (21 amino acids in rodents) that plays a role in the recruitment of the ATP-dependent SWI/SNF chromatin remodeling complex [26] and in redox regulation of C/EBPβ activity [27]. The fact that LAP* has been observed to be expressed in normal human mammary epithelial cells and absent in cancerous mammary epithelial cells although LAP shows exactly the opposite distribution suggests functional consequences for the differential expression of these two C/EBPβ isoforms [28].

C/EBPβ has been characterized as a regulator of differentiation in several systems. C/EBPβ acts in the cascade of events leading to adipocyte differentiation [29], [30]. C/EBPβ has also been shown to have critical roles in the differentiation of the female reproductive system and mammary gland. Female C/EBPβ-deficient mice are sterile due to a block in granulosa cell differentiation [31] and are defective in mammary gland development [32], [33]. Interestingly, pancreatic cells transdifferentiate into hepatocytes when C/EBPβ is overexpressed [34].

Although these studies suggest roles for C/EBPβ in differentiation, C/EBPβ has been associated with proliferation in two recent studies. C/EBPβ augments Ha-ras-induced transformation of NIH3T3 cells and has a critical role in ras-mediated tumorigenesis of keratinocytes [35]. C/EBPβ can also transform a normal mammary epithelial cell line, conferring properties normally associated with neoplastic cells [36].

A growing literature suggests important roles for C/EBP family members in growth and differentiation of osteoblasts. In primary rat osteoblast cultures, C/EBPβ and C/EBPδ are abundantly expressed in proliferating cells, decline during the mid-stage of differentiation, and then subsequently increase at late stages of differentiation [37]. Both C/EBPβ and C/EBPδ are implicated in the regulation of IGF-1 [38] and osteocalcin [37] in osteoblasts. IGF-1 acts as a growth and differentiation factor for bone (reviewed in Ref. [39]), although osteocalcin expression is associated with fully differentiated osteoblasts (reviewed in Ref. [40]). C/EBPβ and C/EBPδ can also act synergistically with Runx2/Cbfa1, a transcription factor and nuclear matrix binding protein (reviewed in [41], [42]), to regulate osteocalcin expression [37]. Runx2/Cbfa1 can in turn upregulate further C/EBPδ expression in osteoblasts [43]. The association of C/EBPβ and δ function with Runx2/Cbfa1 presents a critical linkage between these C/EBP factors and osteoblast differentiation given that Runx2/Cbfa1 is important for osteoblast responsiveness to extracellular matrix signals and is required for expression of genes associated with osteoblast differentiation [44]. Mice with a null mutation of runx2/cbfa1 have a complete absence of bone [45] and humans with cleidocranial dysplasia (CCD, marked by short stature and bone hypoplasia) have been demonstrated to have deletions within the human runx2/cbfa1 gene [46].

In addition to regulating growth and differentiation, C/EBPβ is most well known for its ability to be an effector in the induction of genes responsive to LPS, IL-1, or IL-6 stimulation (reviewed in Ref. [6]). Osteoblasts express C/EBP-responsive genes, including IL-1β, IL-6, IL-8, MCP-1, and TNFα  ([2], [47], [48], [49], [50], reviewed in Ref. [51]). In addition, these cytokines can, in a positive feedback loop, further induce C/EBPβ mRNA levels [2]. In osteoblasts, C/EBPβ has been shown to be induced by cytokines such as IL-1 [52] and is implicated in the regulation of IL-6 transcription [53]. IL-6 is particularly important because it plays a critical role in bone homeostasis and is produced by osteoblasts basally and at enhanced levels in response to known inducers. IL-6 is an important activator of osteoclast activity. Factors that modulate bone resorption such as parathyroid hormone (PTH), PTH-related peptide, androgens, estrogens, and thyroid hormones are also known to correspondingly influence the expression of IL-6. Knockout of IL-6 gene expression in transgenic mice prevented bone loss after oophorectomy [54], further suggesting an important role for IL-6 in inducing bone resorption.

The clear involvement of C/EBP family members in the differentiation and differentiated function of osteoblasts led us to further examine their activity in the temporal context of differentiating MC-3T3-E1 cells. In this report, we have found downregulation of C/EBP DNA-binding activity with mouse osteoblast differentiation. C/EBPβ was found to be the major component of C/EBP DNA-binding activity in proliferating MC3T3-E1 osteoblasts, although differentiating cells displayed greatly reduced C/EBP DNA-binding activity as well as reduced C/EBPβ protein levels. Furthermore, MC3T3-E1 cells stably expressing C/EBPβ LAP from a retroviral vector displayed enhanced proliferation and correspondingly suppressed differentiation as indicated by decreased alkaline phosphatase activity, a marker of osteoblast differentiation.

Section snippets

Cell culture

Cloned osteoblast-like MC3T3-E1 cells derived from newborn mouse calvaria were maintained in α-MEM medium supplemented with 10% fetal bovine serum at 37°C in a humidified atmosphere of 5% CO2/95% air. Stable transductants were maintained in the aforementioned growth medium supplemented with 250 μg/ml G418 (Invitrogen). The cells were seeded at a density of 105 cells/10 cm plate for electrophoretic mobility shift assays (EMSAs), and at 2 × 104 cells/well in 24-well plates for growth and

C/EBPβ DNA-binding activity is abundant in proliferating MC3T3-E1 cells, but is downregulated upon osteoblast differentiation

Studies demonstrating an important role for C/EBP transcription factors in the expression of various gene products associated with osteogenesis and osteoblast function led us to investigate C/EBP DNA-binding activity over a time course of osteoblast differentiation. We chose to investigate C/EBP activity in MC3T3-E1, a nontransformed clonal mouse osteoblast cell line that undergoes developmental stage specific expression of genes and produces a mineralized matrix analogous to normal diploid

Discussion

The data presented in this paper demonstrate a role for C/EBPβ LAP in promoting a proliferative state in MC3T3-E1 osteoblasts and in suppressing osteogenic differentiation. C/EBPβ expression and, consequently, DNA-binding activity were downregulated upon the induction of differentiation in MC3T3-E1 osteoblasts. The maintenance of C/EBPβ LAP activity, by the introduction of a retroviral expression vector allowing constitutive C/EBPβ LAP expression, both promoted the proliferation of MC3T3-E1

Acknowledgements

The authors thank Regina Irwin for expert technical assistance. This research was funded by a grant to RCS and LRM from the Cancer Center at MSU and grants to LRM from NASA (NAG8-1571) and NIH (DK061184).

References (72)

  • E. Kowenz-Leutz et al.

    A C/EBPβ isoform recruits the SWI/SNF complex to activate myeloid genes

    Mol. Cell

    (1999)
  • W.C. Su et al.

    Differential activation of a C/EBPβ isoform by redox switch may confer the lipopolysaccharide inducible expression of IL-6 gene

    J. Biol. Chem.

    (2003)
  • T.L. McCarthy et al.

    Runt domain factor (Runx)-dependent effects on CCAAT/enhancer-binding protein δ expression and activity in osteoblasts

    J. Biol. Chem.

    (2000)
  • V. Geoffroy et al.

    A PEBP2 alpha/AML-1-related factor increases osteocalcin promoter activity through its binding to an osteoblast-specific cis-acting element

    J. Biol. Chem.

    (1995)
  • P. Ducy et al.

    Osf2/Cbfa1: a transcriptional activator of osteoblast differentiation

    Cell

    (1997)
  • S. Mundlos et al.

    Mutations involving the transcription factor CBFA1 cause cleidocranial dysplasia

    Cell

    (1997)
  • S. Natsuka et al.

    Macrophage differentiation-specific expression of NF-IL6, a transcription factor for interleukin-6

    Blood

    (1992)
  • C.L. Cepko et al.

    Construction and applications of a highly transmissible murine retrovirus shuttle vector

    Cell

    (1984)
  • R. Mann et al.

    Construction of a retrovirus packaging mutant and its use to produce helper-free defective retrovirus

    Cell

    (1983)
  • S. Parkin et al.

    Regulation of C/EBP activator proteins by heterodimerization with C/EBPγ (Ig/EBP)

    J. Biol. Chem.

    (2002)
  • N. Franchimont et al.

    Platelet-derived growth factor induces interleukin-6 transcription in osteoblasts through the activator protein-1 complex and activating transcription factor-2

    J. Biol. Chem.

    (1999)
  • Y. Umayahara et al.

    Protein kinase C-dependent, CCAAT/enhancer-binding protein β-mediated expression of insulin-like growth factor I gene

    J. Biol. Chem.

    (2002)
  • D.M. Thomas et al.

    The retinoblastoma protein acts as a transcriptional coactivator required for osteogenic differentiation

    Mol. Cell

    (2001)
  • P.F. Johnson et al.

    CAAT/enhancer binding (C/EBP) proteins

  • S. Akira et al.

    A nuclear factor for IL-6 expression (NF-IL6) is a member of a C/EBP family

    EMBO J.

    (1990)
  • C.J. Chang et al.

    Molecular cloning of a transcription factor, AGP/EBP, that belongs to members of the C/EBP family

    Mol. Cell. Biol.

    (1990)
  • P. Descombes et al.

    LAP, a novel member of the C/EBP gene family, encodes a liver-enriched transcriptional activator protein

    Genes Dev.

    (1990)
  • D.P. Ramji et al.

    The two C/EBP isoforms, IL-6DBP/NF-IL6 and C/EBPδ/NF-IL6β, are induced by IL-6 to promote acute phase gene transcription via different mechanisms

    Nucleic Acids Res.

    (1993)
  • T. Nakajima et al.

    Phosphorylation at threonone-235 by a ras-dependent mitogen-activated protein kinase cascade is essential for transcription factor NF-IL6

    Proc. Natl. Acad. Sci. U. S. A.

    (1993)
  • M. Wegner et al.

    Calcium-regulated phosphorylation within the leucine zipper of C/EBP

    Science

    (1992)
  • C. Trautwein et al.

    Transactivation by NF-IL6/LAP is enhanced by phosphorylation of its activation domain

    Nature

    (1993)
  • C. Trautwein et al.

    Protein kinase A and C site-specific phosphorylations of LAP (NF-IL6) modulate its binding affinity of DNA recognition elements

    J. Clin. Invest.

    (1994)
  • M. Xu et al.

    STAT5-induced Id-1 transcription involves recruitment of HDAC1 and deacetylation of C/EBPβ

    EMBO J.

    (2003)
  • A.L. Welm et al.

    C/EBPα regulates generation of C/EBPβ isoforms through activation of specific proteolytic cleavage

    Mol. Cell. Biol.

    (1999)
  • C.F. Calkhoven et al.

    Translational control of C/EBPα and C/EBPβ isoform expression

    Genes Dev.

    (2000)
  • N.A. Timchenko et al.

    CUG repeat binding protein (CUGBP1) interacts with the 5′ region of C/EBPβ mRNA and regulates translation of C/EBPβ isoforms

    Nucleic Acids Res.

    (1999)
  • Cited by (28)

    • Transcriptional regulation of adipocyte differentiation: A central role for CCAAT/ enhancer-binding protein (C/EBP) β

      2015, Journal of Biological Chemistry
      Citation Excerpt :

      Additionally, overexpression of a dominant-negative C/EBPβ (A-C/EBP) that blocks C/EBPβ DNA binding activity by dimerizing through its leucine zipper (18) also disrupts MCE and adipogenesis in 3T3-L1 cells (19). Intriguingly, C/EBPβ takes part in the proliferation of certain other cell types such as lobuloalveolar cells, osteoblasts, and keratinocytes (20–22), further supporting an important role of C/EBPβ in cell proliferation. To understand how C/EBPβ promotes MCE, a promoter-wide ChIP-on-chip analysis combined with gene expression microarrays was performed to identify the potential target genes of C/EBPβ at the early stage of 3T3-L1 adipocyte differentiation (8).

    • Instruction of mesenchymal cell fate by the transcription factor C/EBPβ

      2012, Gene
      Citation Excerpt :

      The reports described above suggest that C/EBPβ, both the LAP and the LIP isoform, act as enhancers of osteoblast differentiation. However, before commitment, C/EBPβ may function as a transcriptional repressor of Runx2 and of osteoblast differentiation (Iyer et al., 2004; Wiper-Bergeron et al., 2007b). Once osteogenic differentiation is initiated by osteogenic stimuli, Smad3 expression increases, binds to C/EBPβ and abrogates its inhibitory function on Runx2 transcription.

    • CCAAT/enhancer-binding protein-β participates in oxidized LDL-enhanced proliferation in 3T3-L1 cells

      2011, Biochimie
      Citation Excerpt :

      Under our experimental conditions, C/EBPβ -differently from its physiological temporal expression pattern [20,21]- was strongly phosphorylated and remained in the nucleus for several days where it exerted a strong DNA-binding activity. The promitotic activity of C/EBPβ is well known not only in adipose tissue, but also in a number of other cell types such as hepatocytes [25] and osteoblasts [26]. In addition, C/EBPβ appears to be highly expressed in different types of tumors [27,28].

    • Nicotinic modulation of gene expression in osteoblast cells, MG-63

      2011, Bone
      Citation Excerpt :

      Although it is clear that CEBPB controls osteoblast differentiation, it is unclear and controversial whether CEBPB is promotive or suppressive of osteogenesis. It has been reported that over expression of CEBPB stimulates the proliferation of the mouse osteoblastic cell line, MC3T3-E1, but suppresses its osteoblastic differentiation [28]. Thus, up-regulation of CEBPB may lead to bone metabolism processes due to its stimulatory effect on osteoblast proliferation and bone homeostasis.

    • The Cells of Bone

      2006, Dynamics of Bone and Cartilage Metabolism: Principles and Clinical Applications
    View all citing articles on Scopus
    View full text