Open Access

Investigation of transcriptome mechanism associated with osteoporosis explored by microarray analysis

  • Authors:
    • Yan Liu
    • Ying Li
    • Xing Liu
    • Chun-Sheng Wang
  • View Affiliations

  • Published online on: March 6, 2019     https://doi.org/10.3892/etm.2019.7349
  • Pages: 3459-3464
  • Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Microarray data of osteoporosis (OP) were analyzed based on prediction of transcription factors (TFs) or their targets as well as influences of TFs or TF network to uncover key TFs in OP. The microarray data E-GEOD-35956 was downloaded from the GPL570 platform. Differentially expressed genes (DEGs) with logarithm of fold change (|logFC|) >2 and P-value <0.05 were identified between OP samples and normal controls. TF genes were screened from the DEGs based on ITFP, Marbach 2016, TRRUST databases. TF targets were enriched from DEGs using Fisher's exact test. TF targets were selected based on their impact factors. TF targets were chosen from TF network analysis. Finally, key TFs were identified by based on TFs coverage. A total of 300 DEGs were obtained. There were no TF genes screened from the DEGs. In total 165, 87 and 178 TF targets were screened from DEGs respectively based on Fisher's exact test, influence of TFs or TF network analysis. According to the optimal TF set with TFs having maximum coverage of DEGs, 178 TF targets was the most. Thus, the optimal sets of TFs were FOXO1, KLF16, RXRA, RARA, HNF4A, CEBPB, ESR1, SOX8, ZNF219, and SP1. Altogether, these results suggested identified crucial TFs in OP might play a significant role in OP development, showing these key TFs probably would aid in unveiling the underlying molecular mechanisms and may be therapeutic targets, diagnostic or prognostic biomarkers for OP.

Introduction

Osteoporosis (OP) is a common metabolic skeletal disease in the elderly population featured by osteopenia and microstructure, mainly due to the imbalance between bone resorption and bone formation, resulting in increased morbidity and high cost of health care. Current existing therapeutic agents specific to OP are mainly antiresorptive drugs that repress bone resorptive action of osteoclasts. Largely owing to that the mechanisms of excessive bone resorption have been thoroughly explored (1). However, the medicinal efficacy on bone mass and strength of patients remains limited. Thus, it is imperative to address this public health concern, and identify its underlying molecular mechanisms.

The cellular and molecular processes involved in pathophysiology of OP are relatively complicated regulatory networks, involving numerous genes, factors and several pathways. In molecular regulatory networks, it is considered that alterations of gene expression elicit abnormal protein function and pathways turbulence, thus occurrence and development of disease. It is well-known that substantial genes were regulated by transcription factors (TFs), recognized as the master regulators binding to DNA sequences specifically and thereby modulating gene transcription. The transcriptional regulation elicited by TFs is of importance to normal function of the organism, with transcriptional regulation central to cell cycle growth and survive (2), cell differentiation (3), cell adhesion (4) and cell homeostasis (5). However, TF failure leads to nearly 1/3 of human developmental diseases ascribed to TF misregulations (6,7). Thus, TF prediction is a pivotal step to comprehending sophisticated regulatory molecular networks. To date, effective screening methods of TFs related to OP remained largely lacking. OP mediated by osteoblasts is not able to balance bone absorption mediated by osteoclasts, leading to decrease in bone mass and low bone mineral density. Osteoblasts, as bone-forming cells, derive from mesenchymal stem cells (MSCs) which is the source for bone remodeling, besides, the differentiated osteoblasts are decreased from MSCs with aging concerning series of molecular mechanisms (8). However, research on osteoblast action involving underlying mechanisms are relatively ignored.

Therefore, the present study was designed to investigate the mechanistic TFs for OP progression using bioinformatics methods based on gene expression data from the MSC of OP patients. TF targets enrichment analysis was performed for the chosen differentially expressed genes (DEGs). Then, analysis of TF impact factors (IFs) was conducted for DEGs. Moreover, influence of TF network analysis was executed for DEGs. Finally, the obtained TFs based on TF targets prediction and influence of TFs targets or TF network were analyzed comprehensively to achieve the key TFs, which might contribute to future therapy of OP.

Materials and methods

Microarray data collection and preprocessing

The gene expression profile data GSE35956 were extracted from the study of Benisch et al (9), which was based on Gene Expression Omnibus database. Human mesenchymal stem cells (hMSC) of elderly patients (79–94 years) suffering from OP were isolated from femoral heads after low-energy fracture of the femoral neck. A total of 5 OP patients (elderly) and 5 control group (middle-aged) were included. Then, the raw data were normalized and converted into expression values by the robust multi-array average (RMA) algorithm (www.bioconductor.org) and R statistical software (version 3.0.0; R Project for Statistical Computing, http://www.r-project.org/).

DEG analysis

Multiple linear regression package, limma package (10,11) was used to determine the DEGs between the OP patients and normal controls. Multiple testing corrections were performed using the Bayesian method. Only genes with logarithm of fold change (|logFC|) ≥2 and P-value <0.05 were chosen as the DEGs. To ensure the stability of the object in the screening process, all the DEGs were chosen if their number was over 300 with their |logFC| beyond 2 while the top 300 DEGs were chosen if their the number was below 300 with their |logFC| beyond 2.

TF genes or TF target prediction

It is known that each TF, as a protein, has a corresponding regulatory gene, which mainly binds to specific DNA sequences and thus exerts its regulatory role in incomputable biological process. Hence, TF gene was identified first based on mapping DEGs to TF databases including ITFP (12), Tissue-specific regulatory circuits (13), TRRUST (14) databases involved in TFs genes, their downstream regulated genes (target genes) and binding sites. Then, once no TF genes existed, we speculated potential effect of TF by analyzing whether TF targets contained DEGs using Fisher's exact test, which could reflect that the more TF targets were enriched, the more crucial the TFs were. Noticeably, the abundant TF targets appeared due to likelihood that TF targets were widely regulated by TFs such as TFs involved in regulating cell cycle. Thus, such TFs were needed for the following study combined with disease state.

The influence of TF analysis

IFs of TFs were assessed by calculating G-score according to its metric. By comprehensively considering the G-score (the forum was shown as follow), average G-score and numbers of regulated genes, the IF (expressed in P-value) of TF was gained. The lower of this value is, the bigger of TF influence is. According to the P-value, the crucial TFs targets were predicted.

GSx=|LSx|(-log10PSx)
The influence of TF network analysis

Considering the possibility that TFs could form a co-expression relationship with its target genes, to assess the importance of TFs, Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) and the afore-mentioned 3 databases were used to calculate the influence of TFs on local network neighborhood using the weighted sum method (15). Subsequently, G-score of network was calculated and the crucial TF targets were determined by its ranking.

Identification of optimal set of TFs

To select the optimal set of TFs with the greatest combined influence, TFs with maximum coverage of DEGs were determined. Top TFs obtained based on above third methods (TF targets enrichment analysis, TF targets influences analysis, TF network influences analysis) were integrated to obtain the optimal crucial TFs.

Results

Data preprocessing and DEG identification

After data preprocessing, a total of 20,514 genes were obtained from 5 OP patients (elderly) and 5 control groups (middle-aged). A total of 300 DEGs were obtained, namely, LYG2, PPEF1, TTC16, LOC100134368, LOC100505716, CKM, LOC100506272, ADAMTS7, MAB21L2, NR0B1, which were all upregulated in OP group (Table I).

Table I.

The top 10 DEGs.

Table I.

The top 10 DEGs.

Gene namelogFCAveExprtP-valueadj.P.Val
LYG23.8228635464.89860711611.796545813.09E-070.006343212
PPEF12.9679171135.3068814318.7697004054.84E-060.049630867
TTC163.3713973285.5274930538.1989293858.85E-060.054473657
LOC1001343682.8771902745.5387424777.7290463281.49E-050.054473657
LOC1005057163.0986915915.2038482637.6772710351.58E-050.054473657
CKM3.473193986.2479404157.6701124341.59E-050.054473657
LOC1005062722.3688359644.82448627.192555592.78E-050.081533396
ADAMTS71.3728959957.0749016556.95256713.72E-050.092051022
MAB21L23.6541743529.0464811656.7270155784.91E-050.092051022
NR0B11.9110830955.9976742026.5930369725.82E-050.092051022

[i] DEGs, differentially expressed genes; logFC, logarithm of fold change.

TF genes or TF targets prediction

The result of TF genes enrichment analysis suggested that no DEGs were identified as TF genes. In the following, we analyzed whether TF targets were contained in DEGs using Fisher's exact test. As shown in Table II, 165 TFs targets including CKM, CD74, ADAMTS, ADAMTS7, NPAS1 were enriched, correspondingly, the top 10 TFs were WT1, ZBTB7A, CACBP, ZNF281, ZBTB47, ZNF219, PATZ1, TFAP2C, HGS, and KLF16.

Table II.

Top 10 TF enrichment analysis of DEGs.

Table II.

Top 10 TF enrichment analysis of DEGs.

TF_nameTF_P-valueFDR_pnum_genes
WT16.52E-087.48E-05  97
ZBTB7A1.83E-050.007169601  48
CACBP2.16E-050.007169601118
ZNF2812.50E-050.007169601111
ZBTB473.30E-050.00756938     6
ZNF2193.96E-050.00756938100
PATZ10.0001061060.015445541  79
TFAP2C0.0001289680.015445541112
HGS0.0001323630.015445541     5
KLF160.0001533280.015445541107
EGR0.0001573280.015445541127

[i] TFs, transcription factors; DEGs, differentially expressed genes.

The influence of TF analysis

According to the G-score, average G-score, numbers of regulated genes and the P-value of TF comprehensively, 87 TF targets from DEGs were obtained, correspondingly, the top 10 crucial TFs were FKBP8, SP1, LMO4, KLF3, KHDRBS1, ZFPM1, EML3, ZNF580, ZBTB45, and WDTC1 (Table III).

Table III.

Top 10 TFs identified from DEGs based on their influence.

Table III.

Top 10 TFs identified from DEGs based on their influence.

TF_geneG-scoreave_scorenum_genesrank_p
FKBP81.6296601531.62966015310
SP12063.2531050.206263432100030
LMO45.5952141111.39880352840.02001
KLF35.5063575381.37658938440.02405
KHDRBS12.6960283551.34801417820.02983
ZFPM11.3103401621.31034016210.03837
EML31.281087261.2810872610.04572
ZNF5802.5483927381.27419636920.04752
ZBTB452.4775530681.23877653420.05746
WDTC11.1783556621.17835566210.07665

[i] TFs, transcription factors; DEGs, differentially expressed genes.

The influence of TF network analysis

Based on the G-score of network ranking, 178 TF targets were identified from DEGs, correspondingly, the top 10 influential TFs included FOXO1, KLF16, RXRA, RARA, HNF4A, CEBPB, ESR1, SOX8, ZNF219, and SP1 (Table IV).

Table IV.

Top 10 TFs identified from DEGs based on their G-scores of TF network.

Table IV.

Top 10 TFs identified from DEGs based on their G-scores of TF network.

TF_geneG_net_score
FOXO1328.9369128
KLF16323.1693838
RXRA312.8356811
RARA310.4836752
HNF4A300.1282162
CEBPB297.0888622
ESR1291.421492
SOX8291.0030097
ZNF219290.6270745
SP1285.5734732
NFIC283.8264335

[i] TFs, transcription factors; DEGs, differentially expressed genes.

Optimal set of TF identification

Ultimately, the above third methods were integrated with the greatest combined influence to attain the optimal set of TFs with maximum coverage of DEGs. The PPI network analysis based on the G-score of network efficacy is better than others methods due to the number of the 178 TF targets was more than the number of 165 or 87 TF targets, as shown in Fig. 1. Thus, the optimal set of TFs were FOXO1, KLF16, RXRA, RARA, HNF4A, CEBPB, ESR1, SOX8, ZNF219, and SP1.

Discussion

Bioinformatics methods were utilized to identify crucial TFs for clinical OP treatment. The results suggested that 300 DEGs such as upregulated LYG2, PPEF1, TTC16 were gained in OP group compared with control group. A total of 165 TF targets from DEGs were enriched based on TF genes enrichment analysis, 87 TF targets from DEGs were attained by their IF analysis and 178 TF targets from DEGs were achieved by TF network influence analysis. According to the optimal TF set with TFs having maximum coverage of DEGs, 178 TF targets were the most. Thus, the optimal sets of TFs were FOXO1, KLF16, RXRA, RARA, HNF4A, CEBPB, ESR1, SOX8, ZNF219, and SP1. These TFs identified from third bioinformatics methods are possibly capable of aiding in a better understanding of the underlying molecular mechanisms, implying these crucial TFs are likely to be the potential therapy target for OP.

It is well-established that TF, specific to binding to its target gene, thus exerts inhibitory or facilitative role in gene expression, showing a significant part in the multitude of biological processes involved in diseases (1517). Particularly, it is well known that runt-related TF-2 (Runx-2), served as a master member of osteogenic differentiation specific TFs during the early stage of osteogenesis, upregulates the transcription of various mineralized related protein genes in osteoblasts and chondrocytes, and thus promotes these cells to differentiate into osteoblasts (18,19). Hence, Runx-2 is also proved to play a key role in bone metabolism regulation and bone development, indicating it is of great significance to prevent and treat OP. Osterix, a zinc-finger-containing TF, is an essential osteogenic marker for the differentiation of preosteoblasts into mature osteoblasts during the late stage of osteoblast differentiation, which is required for bone formation (20,21). It is reported that nuclear receptors, emerged as a family of TFs, are fundamental regulators of maintaining bone development and remodeling, besides, several drugs targeting it are widely applied in treating bone diseases such OP via regulating rates of bone formation and resorption (22). Although some TFs are proved to play an essential role in bone metabolism, global prediction of TFs in OP are still relatively scarce.

Thus, in the present study, comprehensive bioinformatics methods were introduced to obtain key TFs from a global point of view. First, 300 DEGs were obtained including top 10 DEGs such as LYG2, PPEF1, TTC16 all upregulated in OP according to 5 OP samples compared with 5 normal samples. Following, based on third bioinformatics methods, 165 TF targets, 87 TF targets and 178 TF targets were identified, respectively. The 178 TF targets had the most coverage of TFs, thus, the optimal TF set of these TF targets were FOXO1, KLF16, RXRA, RARA, HNF4A, CEBPB, ESR1, SOX8, ZNF219, and SP1. It is suggested that deletion of forkhead box O1 (FoxO1), one of members of the Forkhead box O (FOXO) family of TFs, could lead to a bone formation increase, which was maintained up to 24 months in mice while a lower number of adipocytes in the bone marrow of Foxo-deleted mice at this late age was presented (23), indicating FoxO1 possibly plays a crucial regulatory role in bone metabolism. It has also been demonstrated that Retinoid-X receptor-α (RXRA), one of nuclear hormone superfamily, is an essential cofactor in the action of 1,25-dihydroxyvitamin D (1,25[OH]2-vitamin D) and epigenetically regulates activation of vitamin D, indicating it may influence bone mineral accrual (2426). It is shown that CCAAT/enhancer-binding protein β (CEBPB), a TF, reduced bone mass in knockout mice (27). It has been found that estrogen receptor α (ESR1) is associated with low mineral osseous densitometry in women after menopause (28). Schmidt et al have shown that Sox8-deficient mouse exhibit a severely impaired bone formation, which is modulated by a strongly reduced expression of runt-related TF 2 (29). Taken together, the above evidence indicates that the comprehensive bioinformatics analysis of TFs from a point of view provided by our study may aid in a better understanding of the molecular mechanism of OP. Albeit a novel insight of comprehending OP molecular pathogenesis emerged, many further studies are required due to the presence of some limitations in this study. Relatively small sample number and datasets, verification of in vitro and in vivo experiments are needed in further investigations.

In conclusion, our study provided novel insight into the mechanism by which crucial TFs were identified by comprehensive bioinformatics analysis of OP microarray data. These selected TFs are possible potential targets in the management of OP.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

CSW conceived the study and drafted the manuscript. YLiu, YLi and XL performed the experiments, analyzed the data and revised the manuscript. All authors read and approved the final study.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Pavone V, Testa G, Giardina SMC, Vescio A, Restivo DA and Sessa G: Pharmacological therapy of osteoporosis: A systematic current review of literature. Front Pharmacol. 8:8032017. View Article : Google Scholar : PubMed/NCBI

2 

Dhruv HD, McDonough Winslow WS, Armstrong B, Tuncali S, Eschbacher J, Kislin K, Loftus JC, Tran NL and Berens ME: Reciprocal activation of transcription factors underlies the dichotomy between proliferation and invasion of glioma cells. PLoS One. 8:e721342013. View Article : Google Scholar : PubMed/NCBI

3 

Cheng D, Wang S, Jia W, Zhao Y, Zhang F, Kang J and Zhu J: Regulation of human and mouse telomerase genes by genomic contexts and transcription factors during embryonic stem cell differentiation. Sci Rep. 7:164442017. View Article : Google Scholar : PubMed/NCBI

4 

Silva LM, Hirai KE, de Sousa JR, de Souza J, Dias LB Jr, Carneiro FRO, Aarão TLS, Fuzii HT and Quaresma JAS: NFκB transcription factor (p65) immunohistochemistry in leprosy dermal microvasculature. Microb Pathog. 113:427–431. 2017. View Article : Google Scholar : PubMed/NCBI

5 

He X, Lindsay-Mosher N, Li Y, Molinaro AM, Pellettieri J and Pearson BJ: FOX and ETS family transcription factors regulate the pigment cell lineage in planarians. Development. 144:4540–4551. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Heppler LN and Frank DA: Targeting oncogenic transcription factors: Therapeutic implications of endogenous STAT inhibitors. Trends Cancer. 3:816–827. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Ruetz T, Pfisterer U, Di Stefano B, Ashmore J, Beniazza M, Tian TV, Kaemena DF, Tosti L, Tan W, Manning JR, et al: Constitutively active SMAD2/3 are broad-scope potentiators of transcription-factor-mediated cellular reprogramming. Cell Stem Cell. 21:791–805.e9. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Garg P, Mazur MM, Buck AC, Wandtke ME, Liu J and Ebraheim NA: Prospective review of mesenchymal stem cells differentiation into osteoblasts. Orthop Surg. 9:13–19. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Benisch P, Schilling T, Klein-Hitpass L, Frey SP, Seefried L, Raaijmakers N, Krug M, Regensburger M, Zeck S, Schinke T, et al: The transcriptional profile of mesenchymal stem cell populations in primary osteoporosis is distinct and shows overexpression of osteogenic inhibitors. PLoS One. 7:e451422012. View Article : Google Scholar : PubMed/NCBI

10 

Diboun I, Wernisch L, Orengo CA and Koltzenburg M: Microarray analysis after RNA amplification can detect pronounced differences in gene expression using limma. BMC Genomics. 7:2522006. View Article : Google Scholar : PubMed/NCBI

11 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

12 

Zheng G, Tu K, Yang Q, Xiong Y, Wei C, Xie L, Zhu Y and Li Y: ITFP: An integrated platform of mammalian transcription factors. Bioinformatics. 24:2416–2417. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Marbach D, Lamparter D, Quon G, Kellis M, Kutalik Z and Bergmann S: Tissue-specific regulatory circuits reveal variable modular perturbations across complex diseases. Nat Methods. 13:366–370. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Han H, Shim H, Shin D, Shim JE, Ko Y, Shin J, Kim H, Cho A, Kim E, Lee T, et al: TRRUST: A reference database of human transcriptional regulatory interactions. Sci Rep. 5:114322015. View Article : Google Scholar : PubMed/NCBI

15 

Szklarczyk D, Franceschini A, Kuhn M, Simonovic M, Roth A, Minguez P, Doerks T, Stark M, Muller J, Bork P, et al: The STRING database in 2011: Functional interaction networks of proteins, globally integrated and scored. Nucleic Acids Res. 39:(Database). D561–D568. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Wilkinson AC, Nakauchi H and Göttgens B: Mammalian transcription factor networks: Recent advances in interrogating biological complexity. Cell Syst. 5:319–331. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Cohen-Solal KA, Kaufman HL and Lasfar A: Transcription factors as critical players in melanoma invasiveness, drug resistance, and opportunities for therapeutic drug development. Pigment Cell Melanoma Res. 31:241–252. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Thu HE, Mohamed IN, Hussain Z and Shuid AN: Exploring molecular mechanism of bone-forming capacity of Eurycoma longifolia: Evidence of enhanced expression of bone-related biomarkers. J Ayurveda Integr Med. S0975-9476(17)30007-4. 2017.PubMed/NCBI

19 

Zhao XL, Chen JJ, Zhang GN, Wang YC, Si SY, Chen LF and Wang Z: Small molecule T63 suppresses osteoporosis by modulating osteoblast differentiation via BMP and WNT signaling pathways. Sci Rep. 7:103972017. View Article : Google Scholar : PubMed/NCBI

20 

Zhang C: Transcriptional regulation of bone formation by the osteoblast-specific transcription factor Osx. J Orthop Surg Res. 5:372010. View Article : Google Scholar : PubMed/NCBI

21 

Choi YH, Han Y, Jin SW, Lee GH, Kim GS, Lee DY, Chung YC, Lee KY and Jeong HG: Pseudoshikonin I enhances osteoblast differentiation by stimulating Runx2 and Osterix. J Cell Biochem. 119:748–757. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Zuo H and Wan Y: Nuclear receptors in skeletal homeostasis. Curr Top Dev Biol. 125:71–107. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Iyer S, Ambrogini E, Bartell SM, Han L, Roberson PK, de Cabo R, Jilka RL, Weinstein RS, O'Brien CA, Manolagas SC, et al: FOXOs attenuate bone formation by suppressing Wnt signaling. J Clin Invest. 123:3409–3419. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Takeyama K and Kato S: The vitamin D3 1alpha-hydroxylase gene and its regulation by active vitamin D3. Biosci Biotechnol Biochem. 75:208–213. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Li G, Yin W, Chamberlain R, Hewett-Emmett D, Roberts JN, Yang X, Lippman SM and Clifford JL: Identification and characterization of the human retinoid X receptor alpha gene promoter. Gene. 372:118–127. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Zhang M, Zhao LJ, Zhou Y, Badr R, Watson P, Ye A, Zhou B, Zhang J, Deng HW, Recker RR, et al: SNP rs11185644 of RXRA gene is identified for dose-response variability to vitamin D3 supplementation: A randomized clinical trial. Sci Rep. 7:405932017. View Article : Google Scholar : PubMed/NCBI

27 

Brommage R, Liu J, Hansen GM, Kirkpatrick LL, Potter DG, Sands AT, Zambrowicz B, Powell DR and Vogel P: High-throughput screening of mouse gene knockouts identifies established and novel skeletal phenotypes. Bone Res. 2:140342014. View Article : Google Scholar : PubMed/NCBI

28 

Gonçalves CG, Almeida BC, Camargo-Kosugi CM, Costa AM, Silva ID and Haidar MA: Polymorphisms in CYP17, COMT, and ESR1 genes in women after menopause and association with bone mineral density. Genet Mol Res. 14:15802–15810. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Schmidt K, Schinke T, Haberland M, Priemel M, Schilling AF, Mueldner C, Rueger JM, Sock E, Wegner M and Amling M: The high mobility group transcription factor Sox8 is a negative regulator of osteoblast differentiation. J Cell Biol. 168:899–910. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2019
Volume 17 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu Y, Li Y, Liu X and Wang C: Investigation of transcriptome mechanism associated with osteoporosis explored by microarray analysis. Exp Ther Med 17: 3459-3464, 2019
APA
Liu, Y., Li, Y., Liu, X., & Wang, C. (2019). Investigation of transcriptome mechanism associated with osteoporosis explored by microarray analysis. Experimental and Therapeutic Medicine, 17, 3459-3464. https://doi.org/10.3892/etm.2019.7349
MLA
Liu, Y., Li, Y., Liu, X., Wang, C."Investigation of transcriptome mechanism associated with osteoporosis explored by microarray analysis". Experimental and Therapeutic Medicine 17.5 (2019): 3459-3464.
Chicago
Liu, Y., Li, Y., Liu, X., Wang, C."Investigation of transcriptome mechanism associated with osteoporosis explored by microarray analysis". Experimental and Therapeutic Medicine 17, no. 5 (2019): 3459-3464. https://doi.org/10.3892/etm.2019.7349