Elsevier

Biomaterials

Volume 35, Issue 23, July 2014, Pages 6015-6025
Biomaterials

The epigenetic promotion of osteogenic differentiation of human adipose-derived stem cells by the genetic and chemical blockade of histone demethylase LSD1

https://doi.org/10.1016/j.biomaterials.2014.04.055Get rights and content

Abstract

Human adipose-derived stem cells (hASCs) are a highly attractive source in bone tissue engineering. It has become increasingly clear that chromatin regulators play an important role in cell fate determination. However, how osteogenic differentiation of hASCs is controlled by epigenetic mechanisms is not fully understood. Here we use genetic tools and chemical inhibitors to modify the epigenetic program of hASCs and identify lysine-specific demethylase 1 (LSD1), a histone demethylase that specifically catalyzes demethylation of di- and mono- methyl histone H3 lysine 4 (H3K4me2/1), as a key regulator in osteogenic differentiation of hASCs. Specifically, we demonstrated that genetic depletion of LSD1 with lentiviral strategy for gene knockdown promoted osteogenic differentiation of hASCs by cell studies and xenograft assays. At the molecular level, we found that LSD1 regulates osteogenesis-associated genes expression through its histone demethylase activity. Significantly, we demonstrated LSD1 demethylase inhibitors could efficiently block its catalytic activity and epigenetically boost osteogenic differentiation of hASCs. Altogether, our study defined the functional and biological roles of LSD1 and extensively explored the effects of its enzymatic activity in osteogenic differentiation of hASCs. A better understanding of how LSD1 influences on osteogenesis associated epigenetic events will provide new insights into the modulation of hASCs based cell therapy and improve the development of bone tissue engineering with epigenetic intervention.

Introduction

Human adipose-derived stem cells (hASCs), one type of mesenchymal stem cells, possess a high plasticity to differentiate into multiple lineages including cartilage, bone, muscle and adipose in the presence of appropriate signaling factors and culture conditions [1], [2], [3], [4], [5]. Currently, hASCs have become a highly attractive source in tissue engineering and cell-based therapy of damaged bone defects, spinal fusion and skeletal reconstruction bone [2], [6], [7]. The main reason lies in the fact that hASCs can be obtained easily from adipose tissues carrying a more abundant and accessible pool of mesenchymal stem cells with a less invasive and less expensive procedures [8], [9], [10].

The critical issue for application of stem cells in tissue engineering is the initiation and control of cellular differentiation in a precise and appropriate manner. Recent evidence suggests that epigenetic regulation including DNA methylation and histone modification plays a key role in fate maintenance and lineage commitment of embryonic stem cells as well as mesenchymal stromal/stem cells [11], [12], [13], [14], [15], [16]. Slight variations of these epigenetic components might result in the changes of local chromatin configuration or nuclear architecture, impose flexible but precise control over the expression of the important regulatory genes, and eventually influence on cell fate determination without changes of the DNA sequences. Unlike genetic alterations, epigenetic changes are reversible and accessible to be regulated, and as such, drugs that target critical epigenetic regulators in cell commitment or differentiation are being overwhelmingly investigated [17], [18].

Lysine-specific demethylase 1 (LSD1/KDM1A) is a flavin adenine dinucleotide (FAD)-dependent amine oxidase that catalyzes mono- and di-methyl moieties removal from histone H3 lysine 4 (H3K4) [19], [20], [21]. Despite progress in understanding the dynamic histone methylation regulation and in revealing the diverse molecular interactions for LSD1, the biological function of LSD1 is just beginning to be uncovered. Recent studies have linked LSD1 to certain high-risk tumors [22], [23], [24], [25], [26], [27], [28], [29], [30]. Moreover, LSD1 has been identified as one of the chromatin regulators implicated in the control of early embryogenesis [15], [31], [32], [33]. In addition, it was reported that LSD1 represses hematopoietic stem and progenitor cell signatures during blood cell maturation [34]. Indeed, within the framework of the so-called epigenetic therapies, there is a growing interest in LSD1 as a potential drug target [26], [35], [36], [37], [38]. However, whether histone methylation associated epigenetic events impacted by LSD1 and currently developed LSD1 inhibitors can contribute to mesenchymal stem cells differentiation as well as bone tissue engineering are largely unknown.

Here, our study focused on investigating the functional role and the molecular mechanism of histone H3K4 demethylase LSD1, especially its catalytic activity in osteogenic differentiation of hASCs.

Section snippets

Cell culture, osteogenic induction and LSD1 inhibitors

Human adipose-derived stem cells (hASCs) were purchased from ScienCell Research Laboratories (San Diego, CA). Stem cells from 3 donors with different lot numbers of the third passage were used for the in vitro and in vivo experiments. All cell-based in vitro experiments were repeated in triplicate. Osteogenic differentiation was induced with Dulbecco's modified Eagle medium (DMEM) containing 10% fetal bovine serum, 100 IU/ml penicillin/streptomycin, 100 nm dexamethasone, 0.2 mm ascorbic acid

The effect of LSD1 depletion on osteogenic differentiation of hASCs in vitro

Previously, we showed that histone demethylase RBP2 (also known as KDM5A or JARID1A) suppresses the osteogenic potential of hASCs by modulating H3K4me3 status of key osteogenic genes [12]. To further explore the biological function of histone demethylase and to investigate the importance of the regulation of histone H3K4 methylation in bone tissue engineering field, the effect of histone demethylase LSD1 depletion on osteogenic differentiation of hASCs was first examined. The siRNA sequence

Discussion

Here, we provide a strategy with genetic as well as epigenetic intervention to regulate osteogenic differentiation of hASCs. To characterize the function of histone demethylase LSD1 in osteogenic differentiation of hASCs, a combination of in vitro experiments were carried out. By quantifying ALP activity, matrix mineralization capacity and osteogenic gene expression profiles, which are common methods used in osteogenic differentiation studies, we found that LSD1 depletion could promote

Conclusions

In this work, we demonstrated that histone demethylase LSD1 inhibits osteogenic differentiation of hASCs in vitro and in vivo. Moreover, we found that LSD1 regulates the expression of osteogenesis associated genes largely dependent on its catalytic activity. Interestingly, we revealed that LSD1 inhibitors, especially CBB1007 could be used to efficiently promote osteogenic differentiation of hASCs.

Acknowledgments

This work was supported by grants (81200763 to W.G., 81070809 to Y.Z., 81272284 and 91219102 to L.S) from the National Natural Science Foundation of China, grants from Program for New Century Excellent Talents (NCET) in University from Ministry of Education of China to L.S. (NCET-13-0934) and Y.Z. (NCET-11-0026), a grant from PKU School of Stomatolgy for talented young investigators to W.G and construction program for national key clinical specialty from National Health and Family Planning

References (56)

  • Y. Yu et al.

    High expression of lysine-specific demethylase 1 correlates with poor prognosis of patients with esophageal squamous cell carcinoma

    Biochem Biophys Res Commun

    (2013)
  • P. Maroni et al.

    Chemical and genetic blockade of HDACs enhances osteogenic differentiation of human adipose tissue-derived stem cells by oppositely affecting osteogenic and adipogenic transcription factors

    Biochem Biophys Res Commun

    (2012)
  • P.A. Zuk et al.

    Human adipose tissue is a source of multipotent stem cells

    Mol Biol Cell

    (2002)
  • B. Levi et al.

    Human adipose-derived stromal cells stimulate autogenous skeletal repair via paracrine Hedgehog signaling with calvarial osteoblasts

    Stem Cells Dev

    (2011)
  • F. De Toni et al.

    Human adipose-derived stromal cells efficiently support hematopoiesis in vitro and in vivo: a key step for therapeutic studies

    Stem Cells Dev

    (2011)
  • K.C. Hicok et al.

    Human adipose-derived adult stem cells produce osteoid in vivo

    Tissue Eng

    (2004)
  • L. Hong et al.

    Adipose tissue engineering by human adipose-derived stromal cells

    Cells Tissues Organs

    (2006)
  • B. Levi et al.

    Human adipose derived stromal cells heal critical size mouse calvarial defects

    PLoS One

    (2010)
  • T. Rada et al.

    Adipose tissue-derived stem cells and their application in bone and cartilage tissue engineering

    Tissue Eng Part B Rev

    (2009)
  • H. Tapp et al.

    Adipose-derived stem cells: characterization and current application in orthopaedic tissue repair

    Exp Biol Med (Maywood)

    (2009)
  • M.T. Chung et al.

    Isolation of human adipose-derived stromal cells using laser-assisted liposuction and their therapeutic potential in regenerative medicine

    Stem Cells Transl Med

    (2013)
  • A. Doi et al.

    Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts

    Nat Genet

    (2009)
  • W. Ge et al.

    Inhibition of osteogenic differentiation of human adipose-derived stromal cells by retinoblastoma binding protein 2 repression of RUNX2-activated transcription

    Stem Cells

    (2011)
  • K.M. Sinha et al.

    Regulation of the osteoblast-specific transcription factor Osterix by NO66, a Jumonji family histone demethylase

    EMBO J

    (2010)
  • W.A. Whyte et al.

    Enhancer decommissioning by LSD1 during embryonic stem cell differentiation

    Nature

    (2012)
  • C. Harrison

    Epigenetic drugs: new modulators of readers and erasers

    Nat Rev Drug Discov

    (2013)
  • J. Kaiser

    Epigenetic drugs take on cancer

    Science

    (2010)
  • J.S. Williams et al.

    Lysine-specific demethylase 1: an epigenetic regulator of salt-sensitive hypertension

    Am J Hypertens

    (2012)
  • Cited by (0)

    1

    These authors contributed equally to this work.

    View full text