Elsevier

Bone

Volume 45, Issue 2, August 2009, Pages 367-376
Bone

TNFα promotes osteogenic differentiation of human mesenchymal stem cells by triggering the NF-κB signaling pathway

https://doi.org/10.1016/j.bone.2009.04.252Get rights and content

Abstract

Mesenchymal stem cells are multipotent cells able to differentiate into different mesenchymal lineages. Studies in the past had suggested that two of these mesenchymal differentiation directions, the chondrogenic and the myogenic differentiation, are negatively regulated by the transcription factor NF-κB. Although osteogenic differentiation has been extensively studied, the influence of NF-κB on this differentiation lineage was not subject of detailed analyses in the past. We have analyzed the consequences of TNF-α treatment and genetic manipulation of the NF-κB pathway for osteogenic differentiation of hMSCs. Treatment of hMSCs during differentiation with TNF-α activates NF-κB and this results in enhanced expression of osteogenetic proteins like bone morphogenetic protein2 (BMP-2) and alkaline phosphatase (ALP). In addition, enhanced matrix mineralization was observed. The direct contribution of the NF-κB pathway was confirmed in cells that express a constitutively active version of the NF-κB-inducing kinase IKK2 (CA-IKK2). The IKK2/NF-κB-induced BMP-2 up-regulation results in the enhancement of RUNX2 and Osterix expression, two critical regulators of the osteogenic differentiation program. Interestingly, a genetic block of the NF-κB pathway did not interfere with osteogenic differentiation. We conclude that TNFα mediated NF-κB activation, although not absolutely required for BMP-2 expression and matrix mineralization nevertheless supports osteogenic differentiation and matrix mineralization by increasing BMP-2 expression. Our results therefore suggest that NF-κB activation may function in lineage selection during differentiation of hMSCs by fostering osteogenic differentiation at the expense of other differentiation lineages.

Introduction

Adult mesenchymal stem cells (MSC) are derived from bone marrow stroma or connective tissue and can differentiate into various lineages including fibroblasts, osteoblasts, adipocytes, and chondrocytes [11], [15], [57]. The multi-lineage differentiation potential of MSC populations has been extensively studied and culturing conditions for in vitro differentiation have been established. Although much progress has been made regarding the distinct differentiation processes in the last years, the signaling pathways involved in differentiation, are not yet completely understood.

The NF-κB signaling pathway is long known to play an important role in inflammation and control of the immune system [9], [24], [47]. In addition, it regulates the transcription of genes involved in cell growth and cell death [5], [35]. NF-κB belongs to the Rel family of transcription factors and in mammals is encoded by five genes named relA, relB, c-rel, nf-κb1, and nf-κb2. All NF-κB proteins contain a conserved Rel homology domain, which is responsible for DNA binding, dimerization, and interaction with IκB proteins. In their inactive state, NF-κB proteins are located in the cytoplasm as homo- or heterodimers, bound to IκB family proteins, most importantly the IκBα protein. Upon stimulation by e.g. pro-inflammatory cytokines, such as TNF-α, the classical pathway is induced resulting in activation of the IκB-kinase-complex (IKK). This IKK-complex consists of two protein kinases (IKK1/α and IKK2/β) and a regulatory protein NEMO/IKKγ. IKK2 is largely responsible for the IκBα phosphorylation in the classical pathway. This phosphorylation triggers poly-ubiquitination and subsequent degradation of IκBα by the proteasome. NF-κB then translocates into the nucleus where it binds to specific sequences in the regulatory regions of target genes. Through a negative feedback loop newly synthesized IκBα binds to nuclear NF-κB and exports it back to the cytoplasm [9]. In addition to the classical pathway there is also an alternative pathway, which plays a central role in expression of genes involved in development and maintenance of secondary lymphoid organs. This alternative pathway is mainly stimulated via LTβR, BAFFR and CD40 leading to activation of NIK and subsequent activation of IKK1 [67].

A specific role of NF-κB in differentiation of MSC derivatives had been implicated in the past. It was shown that NF-κB induces degradation of the mRNA encoding the myogenic transcription factor MyoD in TNF-α treated myocytes [29], [60]. In addition, NF-κB is responsible for TNF-α-induced muscle protein degradation in differentiated muscle cells [41], [43], [46]. Furthermore, NF-κB influences the chondrogenic differentiation as TNF-α-induced NF-κB was shown to down-regulate mRNA levels of the chondrogenic transcription factor Sox9, thereby inhibiting differentiation of chondrocytes [53, [60]. However, there is also evidence that NF-κB can function as a positive regulator of mesenchymal cell differentiation as it was shown that NF-κB p65 expressed in growth plate chondrocytes facilitates growth plate chondrogenesis and longitudinal bone growth by inducing BMP-2 expression and activity [66].

Less is known about the role of NF-κB signaling in osteoblast differentiation. A negative regulation of osteoblast differentiation by NF-κB was suggested as inhibition of NF-κB signaling activity in osteosarcoma cells (Saos2) results in induction of several osteogenic markers, like BMP-4 and 7, Cbfa1, alkaline phosphatase, osteopontin and osteocalcin [4]. This negative role of NF-κB was also demonstrated by the H2O2-induced, NF-κB-dependent reduction of osteogenic differentiation markers, like alkaline phosphatase, collagen I, and Cbfa1 in rabbit bone marrow stromal cells and calvarial osteoblasts [6]. However, NF-κB was not analyzed directly in these cases, but rather the consequences of stimuli were studied, which amongst other pathways also induce NF-κB. Indeed some inhibiting effects of TNF-α on differentiation processes were documented to be independent of NF-κB signaling. Furthermore, previous studies demonstrated that TNF-α-induced inhibition of the terminal adipogenic differentiation of pre-adipocytes [68] and inhibition of osteogenic differentiation of pre-osteoblastic cells [25], [49] is NF-κB-independent.

To elucidate the role of NF-κB in osteogenesis, we have analyzed its influence on osteogenic differentiation by infecting human mesenchymal stem cells with different NF-κB modulators. Our findings indicate that enhanced NF-κB activity in human mesenchymal stem cells increases osteogenic differentiation, whereas decreased NF-κB signaling does not impede the osteogenic differentiation.

Section snippets

Cell culture

Human mesenchymal stem cells (hMSCs) were established from bone marrow samples with informed consent of the donors and following the guidelines of the ethics committee of the University of Ulm as described previously [22]. hMSCs were cultured in DMEM (GibcoBRL Life Technologies) supplemented with 10% heat inactivated fetal bovine serum (Biochrom AG) (FBS), 1% l-Glutamin (Biochrom AG) and 1% Pen/Strep (Biochrom AG) (growth medium) in a humidified atmosphere of 5% CO2 in air. The technique of

TNF-α increases extracellular matrix mineralization of osteogenic differentiated human mesenchymal stem cells

It had been shown that TNF-α-induced NF-κB inhibits mesenchymal cell differentiation into the myogenic and chondrogenic direction by down-regulating the critical transcription factors MyoD and Sox9, respectively [29], [60]. We wanted to determine the influence of the TNF-α/NF-κB-system on the osteogenic differentiation of human mesenchymal stem cells. In osteogenic differentiation, matrix mineralization is an essential hallmark. In this process calcium and phosphate are deposited as

Discussion

Our data show that TNF-α promotes osteogenic differentiation of human mesenchymal stem cells by activating the NF-κB signaling pathway. These effects of TNF-α and NF-κB-induction were monitored by the increased deposition of calcium to the extracellular matrix and the enhanced expression of the osteoinductive growth factor BMP-2 and the osteogenic marker ALP. The observation that elevated NF-κB activity in hMSCs infected with the constitutively active IKK2 also leads to enhanced expression of

Acknowledgments

We would like to thank Dr. Ralf Marienfeld, Dr. Bernd Baumann, and Kay Klapproth for reading the manuscript and for many helpful comments.

This work was supported by grants from BMBF (01GN0123) and the Fonds der Chemischen Industrie to T.W.

References (72)

  • HuberM.A. et al.

    The IKK-2/Ikappa Balpha/NF-kappa B pathway plays a key role in the regulation of CCR3 and eotaxin-1 in fibroblasts. A critical link to dermatitis in Ikappa Balpha-deficient mice

    J. Biol. Chem.

    (2002)
  • KatagiriT. et al.

    The non-osteogenic mouse pluripotent cell line, C3H10T1/2, is induced to differentiate into osteoblastic cells by recombinant human bone morphogenetic protein-2

    Biochem. Biophys. Res. Commun.

    (1990)
  • KomoriT. et al.

    Targeted disruption of Cbfa1 results in a complete lack of bone formation owing to maturational arrest of osteoblasts

    Cell

    (1997)
  • LadnerK.J. et al.

    Tumor necrosis factor-regulated biphasic activation of NF-kappa B is required for cytokine-induced loss of skeletal muscle gene products

    J. Biol. Chem.

    (2003)
  • LeeM.H. et al.

    BMP-2-induced Osterix expression is mediated by Dlx5 but is independent of Runx2

    Biochem. Biophys. Res. Commun.

    (2003)
  • LuX. et al.

    Transcriptional regulation of the osterix (Osx, Sp7) promoter by tumor necrosis factor identifies disparate effects of mitogen-activated protein kinase and NF kappa B pathways

    J. Biol. Chem.

    (2006)
  • MaierH.J. et al.

    Critical role of RelB serine 368 for dimerization and p100 stabilization

    J. Biol. Chem.

    (2003)
  • MurakamiS. et al.

    Potent inhibition of the master chondrogenic factor Sox9 gene by interleukin-1 and tumor necrosis factor-alpha

    J. Biol. Chem.

    (2000)
  • NakashimaK. et al.

    The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation

    Cell

    (2002)
  • OkabeT. et al.

    Regulation of ALP activity by TNF-alpha on human dental pulp

    J. Endod.

    (2006)
  • OttoF. et al.

    Cbfa1, a candidate gene for cleidocranial dysplasia syndrome, is essential for osteoblast differentiation and bone development

    Cell

    (1997)
  • ShahA.K. et al.

    Mechanism of BMP-2 stimulated adhesion of osteoblastic cells to titanium alloy

    Biol. Cell.

    (1999)
  • WuS. et al.

    Nuclear factor-kappa B p65 facilitates longitudinal bone growth by inducing growth plate chondrocyte proliferation and differentiation and by preventing apoptosis

    J. Biol. Chem.

    (2007)
  • XuH. et al.

    Signaling pathways utilized by tumor necrosis factor receptor 1 in adipocytes to suppress differentiation

    FEBS Lett.

    (2001)
  • AdamsJ.C. et al.

    Regulation of development and differentiation by the extracellular matrix

    Development

    (1993)
  • AliA.A. et al.

    Rosiglitazone causes bone loss in mice by suppressing osteoblast differentiation and bone formation

    Endocrinology

    (2005)
  • AndelaB. et al.

    Malignant reversion of a human osteosarcoma cell line, Saos-2, by inhibition of NFkappaB

    Biochem. Biophys. Res. Commun.

    (2002)
  • BaumannB. et al.

    Constitutive IKK2 activation in acinar cells is sufficient to induce pancreatitis in vivo

    J. Clin. Invest.

    (2007)
  • BockerW. et al.

    IKK-2 is required for TNF-alpha-induced invasion and proliferation of human mesenchymal stem cells

    J. Mol. Med.

    (2008)
  • BrunnerC. et al.

    BOB.1/OBF.1 controls the balance of TH1 and TH2 immune responses

    EMBO J.

    (2007)
  • CloverJ. et al.

    Integrin subunit expression by human osteoblasts and osteoclasts in situ and in culture

    J. Cell. Sci.

    (1992)
  • CsiszarA. et al.

    Regulation of bone morphogenetic protein-2 expression in endothelial cells: role of nuclear factor-kappaB activation by tumor necrosis factor-alpha, H2O2, and high intravascular pressure

    Circulation

    (2005)
  • DattaH.K. et al.

    The cell biology of bone metabolism

    J. Clin. Pathol.

    (2008)
  • DignamJ.D. et al.

    Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated mammalian nuclei

    Nucleic Acids Res.

    (1983)
  • FickertS. et al.

    Identification of subpopulations with characteristics of mesenchymal progenitor cells from human osteoarthritic cartilage using triple staining for cell surface markers

    Arthritis Res. Ther.

    (2004)
  • FiedlerJ. et al.

    To go or not to go: migration of human mesenchymal progenitor cells stimulated by isoforms of PDGF

    J. Cell. Biochem.

    (2004)
  • Cited by (221)

    View all citing articles on Scopus
    View full text