Open Access

Identification of long non‑coding RNAs expressed during the osteogenic differentiation of human bone marrow‑derived mesenchymal stem cells obtained from patients with ONFH

  • Authors:
    • Tao Li
    • Ke Xiao
    • Yingxing Xu
    • Yuanzhong Ren
    • Yingzhen Wang
    • Haining Zhang
    • Xisheng Weng
    • Yaping Jiang
  • View Affiliations

  • Published online on: August 31, 2020     https://doi.org/10.3892/ijmm.2020.4717
  • Pages: 1721-1732
  • Copyright: © Li 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

Long non‑coding RNAs (lncRNAs) are crucial for the occurrence and development of numerous diseases. Although lncRNAs are involved in the biological activities of stem cells and play crucial roles in stem cell differentiation, the expression of specific lncRNAs during human bone marrow‑derived mesenchymal stem cell (hBMSC) osteogenic differentiation in osteonecrosis of the femoral head (ONFH) and their regulatory roles have not yet been fully elucidated. To the best of our knowledge, the present study is the first to characterize lncRNA expression profiles during hBMSC osteogenic differentiation in ONFH using microarray analysis and RT‑qPCR to confirm the microarray data. A total of 24 downregulated and 24 upregulated lncRNAs were identified and the results of RT‑qPCR were found to be consistent with those of microarray analysis. Bioinformatics analyses, using the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases, were conducted to explore the possible mechanisms and identify the signaling pathways that the lncRNAs are involved in. GO analysis revealed significant changes in the intracellular organelle, Ras protein signal transduction and transferase activity. KEGG pathway analysis revealed that the lncRNAs were closely associated with fatty acid metabolism, apoptosis and the TGF‑β signaling pathway. The overexpression of MAPT antisense RNA 1 (MAPT‑AS1) was found to promote osteogenesis and inhibit the adipogenesis of hBMSCs at the cellular and mRNA levels. On the whole, the findings of the present study identified the lncRNAs and their roles in hBMSCs undergoing osteogenic differentiation in ONFH and provide a new perspective for the pathogenesis of ONFH.

Introduction

As a pathological state with multiple possible etiologies, osteonecrosis of the femoral head (ONFH), also known as avascular necrosis, results in decreased vascular supply to the subchondral bone of the femoral head, ultimately resulting in osteocyte death and the collapse of the articular surface (1). Several studies have examined the pathogenesis of ONFH, which has been demonstrated to involve the apoptosis of osteoblasts and osteocytes (2,3), adipogenesis (4), venous congestion (5,6) and mutations in the COL2A1 gene (7). However, the specific mechanisms underlying the pathology of ONFH remain poorly understood.

The potential of muscle, cartilage, bone and adipose tissue differentiation has resulted in the use of human bone marrow-derived mesenchymal stem cells (hBMSCs) (8) therapeutically in a clinical setting (9-13). As a key function of hBMSCs, osteogenic differentiation plays a crucial role in the formation and remodeling of bone. Long non-coding RNAs (lncRNAs) are transcripts with a length of >200 nucleotides that do not code for any proteins. The critical roles of lncRNAs in various physiological and pathological processes have been proven (14-18). It has also been demonstrated that lncRNAs may also participate in hBMSC osteogenic differentiation (19). Moreover, the abnormal expression of lncRNAs may lead to the development of diseases due to variations in the osteogenic differentiation capacity of hBMSCs. However, the differential expression profiles of lncRNAs expressed in hBMSCs from patients with ONFH have not yet been fully elucidated. Therefore, the present study aimed to examine the role of lncRNAs expressed during the osteogenic differentiation of abnormal hBMSCs obtained from patients with ONFH.

Materials and methods

Cells and cell culture

Patients who had undergone total hip arthroplasty (THA) due to femoral neck fracture or ONFH were included in the present study, whereas patients who had undergone THA for rheumatoid arthritis, ankylosing spondylitis and other diseases were excluded. In total, 3 patients who had undergone THA for the treatment of femoral neck fracture provided normal bone marrow tissue: All 3 were Chinese; 2 were females, aged 60 and 67 years, while the other patient was a 66-year-old male. ONFH bone marrow tissue was also obtained from another 3 patients, of which 2 were females, aged 55 and 61 years and the other patient was a 58-year-old male, on whom THA had been performed for ONFH. Bone marrow tissue was collected at the Affiliated Hospital of Qingdao University from January, 2018 to May, 2018. The femoral bone marrow tissue samples were used to extract the hBMSCs with density gradient separation, as previously described (20). Bone marrow diluted with an equal volume of PBS was layered over lymphocyte separate medium. The mononuclear cell layer was collected following centrifugation. The hBMSCs extracted were cultured in a stem cell medium in a humidified atmosphere with 5% CO2 at 37°C. The Ethics Committee of the Affiliated Hospital of Qingdao University approved the study, while informed consent was obtained from all participants.

Flow cytometric analysis

Surface antigen markers on the hBMSCs were detected using an Apogee A50-MICRO flow cytometer (Apogee Corporation). The hBMSCs were suspended in PBS at a concentration of approximately 106 cells/ml and washed twice with PBS. Approximately 5x105 cells per 500 µl were incubated and stained with 5 ml of mouse anti-human CD34-fluorescein isothiocyanate (FITC) (560942), CD45-FITC (560976), CD73-FITC (561254) and CD90-FITC (561969) antibodies for 20 min at room temperature. All antibodies used were purchased from BD Biosciences.

Osteogenic differentiation of hBMSCs

The hBMSCs (passage 3) were plated in growth medium in 6-well plates. When 80% confluency was reached, mesenchymal stem cell osteogenic differentiation medium was used as the growth medium. The medium was changed every 3 days. The RT-qPCR analysis of osteogenic differentiation markers [alkaline phosphatase (ALP), Runt-related transcription factor 2 (RUNX2), osteopontin (OPN) and bone sialoprotein (BSP)] and staining (ALP staining and Alizarin Red staining) were adopted to detect the osteogenic ability of the hBMSCs. All experiments were performed in triplicate.

RAN extraction and RT-qPCR

Total RNA extraction was performed form the hBMSCs using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.), following instructions provided by the manufacturer. The expression of osteogenesis- and adipogenesis-related genes, including OPN, BSP, Runx2, ALP and peroxisome proliferator-activated receptor γ (PPARγ) were determined by RT-qPCR. Total RNA was reverse transcribed using oligo-dT primers. The cDNA was utilized as a template to amplify target genes with the SYBR Premix Ex Tag kit (Takara Bio, Inc.). The primers of these genes are listed in Table I. Each RNA sample was evaluated in triplicate and PCR cycles were as follows: 94°C for 5 min, 95°C for 30 sec, 58°C for 30 sec, and 72°C for 30 sec (35 cycles), 94°C for 5 min. Relative expression of mRNA was evaluated using the 2-ΔΔCq method and normalized to the expression of GAPDH (21).

Table I

Primers of lncRNAs and the related osteogenic genes.

Table I

Primers of lncRNAs and the related osteogenic genes.

GenePrimer sequence 5'-3'
GAPDH-F GGTCACCAGGGCTGCTTTTA
GAPDH-R GGATCTCGCTCCTGGAAGATG
ALP-F CC ACGTCTTCACATTTGGTG
ALP-R AGACTGCGCCTGGTAGTTGT
OPN-F ACTCGAACGACTCTGATGATGT
OPN-R GTCAGGTCTGCGAAACTTCTTA
RUNX2-F TGTCATGGCGGGTAACGAT
RUNX2-R AAGACGGTTATGGTCAAGGTGAA
BSP-F TGGATGAAAACGAACAAGGCA
BSP-R AAACCCACCATTTGGAGAGGT
PPARγ-F CC TATTGACCCAGAAAGCGATT
PPARγ-R C ATTACGGAGAGATCCACGGA
CEBP-α-F AGGAACACGAAGCACGATCAG
CEBP-α-R C GCACATTCACATTGCACAA
hsa-lncRNA-AC107070.1-F C ACATTCCAGCCAAGGTAG
hsa-lncRNA-AC107070.1-R C AGCCTCTCAGACCACATTC
hsa-lncRNA-linc-ANKRD20A1-4-F TGGAGTTGGACATTTGTGG
hsa-lncRNA-linc-ANKRD20A1-4-R TGGAGTTGGACATTTGTGG
hsa-lncRNA-LINC00473-F GAGGTCTGAGTCCGAAGTTG
hsa-lncRNA-LINC00473-R AGCAGGCAGATTCCAAAG
hsa-lncRNA-MAPT-AS1-F TCCGCTGGAAAGAGAACTC
hsa-lncRNA-MAPT-AS1-R CC TGTGAGGGCATACACC
hsa-lncRNA-RP11-794G24.1-F GGCGTGGATCTTGGAGAGTC
hsa-lncRNA-RP11-794G24.1-R GATGCTGGACGAATCCCAGT
hsa-lncRNA-AP005273.1-F TTCTTGACCCTCTCCAATGTGA
hsa-lncRNA-AP005273.1-R ACTGTCCAATAGCTTCCATCAGG
Staining

Alizarin Red and ALP staining were adopted to evaluate the osteogenic differentiation capacity of the hBMSCs. An ALP staining kit (Tianjin Blood Research Institute) was used on day 3, as instructed by the manufacturer, to conduct ALP staining. The cells are processed according to the following procedures: No. 1 solution was added at room temperature for 1 min, followed by rinsing for 2 min. The staining solution was then added followed by incubation at 37°C for 2 h and rinsing for 2 min. No. 5 solution was then added for re-staining for 5 min, followed by rinsing for 2 min, and drying. For Alizarin Red staining, after washing the cells twice with PBS, fixing was performed using 95% ethanol for 20 min, and the cells were washed 3 times using distilled water, and were stained using Alizarin Red solution (Sigma-Aldrich; Merck KGaA) for 30 min at 37°C. Oil Red O staining was performed to evaluate the adipogenic differentiation capacity of the hBMSCs. Cells were washed twice with PBS and fixed with 10% formalin for 10 min at room temperature. After fixation, cells were stained with filtered Oil Red O solution (Sigma-Aldrich; Merck KGaA) for 1 h at room temperature.

Microarray analysis

Three normal cell samples of osteogenic differentiation were used as the controls, while 3 osteogenic differentiation samples obtained from patients with ONFH were used as the experimental group. TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) was used to extract total RNA from the hBMSCs, while a mirVana miRNA Isolation kit (Ambeon; Thermo Fisher Scientific, Inc.) was used as instructed by the manufacturer for purification. Following RNA extraction, labeling, hybridization and amplification, the CapitalBiotech human array used 4 identical arrays for each slide to design the lncRNA Array V4.0, while each array contained probes that could interrogate approximately 41,000 human lncRNAs. The probes were used to detect each RNA and the process was repeated for confirmation. A total of 4,974 control probes (Agilent Technologies, Inc.) constituted the array. The analysis of the lncRNA array data was conducted using GeneSpring software V13.0 (Agilent Technologies, Inc.) for data summarization, normalization and quality control. The differentially expressed genes were obtained using a t-test P-value of 0.05 and a fold change of ≥2 and ≤-2 as the threshold values. The adjust data function of CLUSTER 3.0 software was applied for log2 transformation of the data and median centering by the genes, while further analysis was conducted using hierarchical clustering with an average linkage.

Using RT-qPCR, the identity of 6 lncRNAs [AC107070.1, linc-ANKRD20A1-4, RP11-794G24.1, long intergenic non-protein coding RNA 473 (LINC00473), MAPT antisense RNA 1 (MAPT-AS1) and AP005273.1] was further confirmed. The primers used for the lncRNAs are listed in Table I.

Furthermore, hBMSCs were islated from another 30 samples, including 15 normal and 15 patients with ONFH who underwent THA in the Department of Joint surgery at The Affiliated Hospital of Qingdao University from October, 2018 to September, 2019. Informed consent form was obtained from all participants. The expression levels of 2 upregulated lncRNAs (AC107070.1 and linc-ANKRD20A1-4) and 2 downregulated lncRNAs (LINC00473 and MAPT-AS1) were examined.

Target gene prediction

In the present study, the functions of cis and transtarget mRNAs were used to predict the target genes of the lncRNAs. Protein-coding genes within a 100 kb genomic distance from the lncRNA were defined as potentially cis-regulated target genes, and protein-coding genes co-expressed with the lncRNA with a Pearson's correlation coefficient (|r|>0.95) and a >100 kb genomic distance from the lncRNA or in different chromosomes were defined as potentially trans-regulated target genes.

Lentivirus vector construction and infection of hBMSCs for MAPT-AS1

The pRLenti-EF1a-EGFP-CMV-MAP T-AS1-overexpression lentivirus (H12785) was obtained from OBiO Technology. The hBMSCs were infected with MAPT-AS1-overexpression lentivirus at a final multiplicity of infection (MOI) of 100 containing 5 µg/ml polybrene, and observed for the expression of green fluorescent protein (GFP) using an inverted fluorescence microscope (EVOS FL, Invitrogen; Thermo Fisher Scientific, Inc.) after 24 h. The transfection efficiency of MAPT-AS1 was determined by RT-qPCR after 3 days.

Bioinformatics analysis

Gene Ontology (GO) was utilized to identify the molecular functions of the differentially expressed genes. The GO category was also calculated. Furthermore, the differentially expressed lncRNAs were used in a pathway analysis that was performed using the latest Kyoto Encyclopedia of Genes and Genomes (KEGG) database to analyze the potential functions of target genes.

Statistical analysis

All statistical analyses were conducted using SPSS statistical software v.16.0 (SPSS, Inc.). All data are expressed as the means ± standard deviation. Comparisons between 2 variables of microarray data was performed using the Student's t-test. Comparisons between multiple groups were performed using the Kruskal-Wallis test along with Dunn's post hoc test. The χ2 test and Fisher's exact test were adopted for the GO and KEGG analyses. Statistical significance was considered to be indicated by P-values of <0.05.

Results

hBMSCs and osteogenic differentiation

The hBMSCs isolated from the bone marrow samples were spindle-shaped cells (Fig. 1) and no morphological differences were found between the 2 groups. ALP staining, Alizarin Red staining and osteogenic markers, including ALP, RUNX2, OPN and BSP, were used to detect the osteogenic ability of the hBMSCs. Positive ALP staining and Alizarin Red staining results revealed mineral deposits and bone formation (Fig. 1). The results of flow cytometric analysis are also shown in Fig. 1. The results of RT-qPCR revealed the elevated expression levels of OPN, RUNX2, ALP and BSP (Fig. 2).

Expression profiles of lncRNAs in hBMSCs from patients with ONFH

lncRNA expression levels during hBMSC osteogenic differentiation were detected using lncRNA microarray chips (The CapitalBiotech human lncRNA Array V4.0). A total of 48 differentially expressed lncRNAs were identified, including 24 lncRNAs, which were upregulated (RP11-216N14.9, RP11-989E6.10, BX571672.1, RP11-230G5.2, linc-SLC30A5-5, BX004987.4, AC107070.1, RP11-265D17.2, RP11-1060J15.4, RP11-794G24.1, linc-ANKRD20A1-4, RP11-485O10.2, AC097532.2, RP11-143J24.1, RP11-762H8.3, CTD-2015H3.2, linc-C17orf97-2, RP11-513M1.1, RP11-22H5.2, linc-ANKRD20A1-2, linc-OR4M2-5, RP11-262H14.1, linc-LOC389493-3 and linc-ATP6V1C2-3) and 24 lncRNAs, which were downregulated (AC104135.3, RP11-26M5.3, MAPT-AS1, AL589743.1, linc-SLITRK1-4, RP11-229P13.19, AP005273.1, RP11-406O23.2, AP003900.6, RP11-525J21.1, linc-PENK-1, linc-PENK-2, RP5-1102E8.3, linc-CSTB-3, CTD-2314B22.3, RP5-1148A21.3, LINC00473, CTC-498M16.2, RP11-16M8.2, AP000525.9, linc-TCF4-3, RP11-645N11.2, RP11-17A4.2 and RP11-324O2.3). The differential expression of the 48 lncRNAs is presented in brief in Table II. Hierarchical clustering analysis revealed the expression profiles of the lncRNAs during the osteogenic differentiation of hBMSCs from patients with ONFH and healthy subjects (Fig. 3).

Table II

Differential expression of the lncRNAs during the osteogenic differentiation of hBMSCs from patients with ONFH.

Table II

Differential expression of the lncRNAs during the osteogenic differentiation of hBMSCs from patients with ONFH.

lncRNA nameRegulationChromosomeStrandstartEndGeneClassDatabase
RP11-216N14.9Up1-153723458153724652 ENSG00000233222.2AntisenseENSEMBL
RP11-989E6.10Up16-3334430833348279 ENSG00000261200.1IntergenicENSEMBL
BX571672.1Up1+143119060143158077 ENSG00000230850.2IntergenicENSEMBL
RP11-230G5.2Up12-6591704265932479 ENSG00000250748.2IntergenicENSEMBL
linc-SLC30A5-5Up5+6780206167807619XLOC_004413Intergenic HumanLincRNACatalog
BX004987.4Up1-143429871143467644 ENSG00000185044.8IntergenicENSEMBL
AC107070.1Up2-40076834021626 ENSG00000237401.2IntergenicENSEMBL
RP11-265D17.2Up11-1228297212284720 ENSG00000254680.1AntisenseENSEMBL
RP11-1060J15.4Up12-2784948827857344 ENSG00000256377.1AntisenseENSEMBL
RP11-794G24.1Up11-6130698761309731 ENSG00000256443.1IntronicENSEMBL
linc-ANKRD20A1-4Up9+6705114467269646XLOC_007374Intergenic HumanLincRNACatalog
RP11-485O10.2Up15+4907538649076318 ENSG00000259670.1AntisenseENSEMBL
AC097532.2Up2-133043367133051950 ENSG00000230803.1IntergenicENSEMBL
RP11-143J24.1Up15-3029764530338051 ENSG00000259647.1IntergenicENSEMBL
RP11-762H8.3Up15-7854988678556498 ENSG00000259708.1AntisenseENSEMBL
CTD-2015H3.2Up18+16551771779956 ENSG00000266450.1IntergenicENSEMBL
linc-C17orf97-2Up17+5620856601XLOC_012065Intergenic HumanLincRNACatalog
RP11-513M1.1Up18+1089361410894803 ENSG00000263952.1IntronicENSEMBL
RP11-22H5.2Up16+8280692382863243 ENSG00000260862.1IntronicENSEMBL
linc-ANKRD20A1-2Up9+6734051667343501XLOC_007377Intergenic HumanLincRNACatalog
linc-OR4M2-5Up15+2050564220531084XLOC_011157Intergenic HumanLincRNACatalog
RP11-262H14.1Up9+6645728466466010 ENSG00000238113.2IntergenicENSEMBL
linc-LOC389493-3Up7-5668391556685484XLOC_006459Intergenic HumanLincRNACatalog
linc-ATP6V1C2-3Up2+1070242010706471XLOC_001343Intergenic HumanLincRNACatalog
AC104135.3Down2+7515536575160151 ENSG00000204792.2IntergenicENSEMBL
RP11-26M5.3Down8+5306337953067452 ENSG00000254314.1IntronicENSEMBL
MAPT-AS1Down17-4392101643972966 ENSG00000264589.1AntisenseENSEMBL
AL589743.1Down14+1965004119718563 ENSG00000225210.5IntergenicENSEMBL
linc-SLITRK1-4Down13-8568597885722284XLOC_010679Intergenic HumanLincRNACatalog
RP11-229P13.19Down9+139869545139871433 ENSG00000238268.2DivergentENSEMBL
AP005273.1Down11+6426832464272858 ENSG00000232500.1IntergenicENSEMBL
RP11-406O23.2Down9-112522639112534323 ENSG00000232939.1IntronicENSEMBL
AP003900.6Down21+1116978711184046 ENSG00000271308.1ENSEMBL
RP11-525J21.1Down4-6063353360657832 ENSG00000249892.1IntergenicENSEMBL
linc-PENK-1Down8-5743237157472069XLOC_007087Intergenic HumanLincRNACatalog
linc-PENK-2Down8-5744795957449765XLOC_007088Intergenic HumanLincRNACatalog
RP5-1102E8.3Down1+7710256177103024 ENSG00000272855.1ENSEMBL
linc-CSTB-3Down21-4522540245230480XLOC_014107Intergenic HumanLincRNACatalog
CTD-2314B22.3Down14-1988380019925345 ENSG00000244306.5IntergenicENSEMBL
RP5-1148A21.3Down6-6428090964282313 ENSG00000266680.1AntisenseENSEMBL
LINC00473Down6-166361710166401536 ENSG00000223414.2IntergenicENSEMBL
CTC-498M16.2Down5-8770566387734907 ENSG00000250156.2IntergenicENSEMBL
RP11-16M8.2Down8-5743287357472243 ENSG00000246430.2IntergenicENSEMBL
AP000525.9Down22-1615882816159470 ENSG00000206195.6ENSEMBL
linc-TCF4-3Down18-5372780653735555XLOC_012851Intergenic HumanLincRNACatalog
RP11-645N11.2Down7+102613968102629303 ENSG00000230257.1IntronicENSEMBL
RP11-17A4.2Down8+5740165657439695 ENSG00000254254.1IntergenicENSEMBL
RP11-324O2.3Down10-114166031114169248 ENSG00000232934.2AntisenseENSEMBL

[i] hBMSCs, human bone marrow-derived mesenchymal stem cells; ONFH, osteonecrosis of the femoral head.

Comparison between the RT-qPCR and microarray analyses

The microarray data analysis revealed 3 upregulated lncRNAs (AC107070.1, linc-ANKRD20A1-4 and RP11-794G24.1) and 3 downregulated lncRNAs (LINC00473, MAPT-AS1 and AP005273.1) which were selected for RT-qPCR analysis in the hBMSCs. The results of RT-qPCR revealed that the expression trends of these 4 lncRNAs were consistent with those of the microarray results, which are shown in Fig. 4.

The expression levels of AC107070.1, linc-ANKRD20A1-4, LINC00473 and MAPT-AS1 (Fig. 5) in the samples were also consistent with those of the results of the microarray analysis.

Target gene prediction and association study

In the present study, the genes involved were predicted based on the functional annotations of their related cisand transtarget mRNAs. Further results are presented in detail in Table III.

Table III

Predicted target genes of differentially expressed lncRNAs.

Table III

Predicted target genes of differentially expressed lncRNAs.

lncRNATarget gene
A, Partial lncRNAs overexpressed in hBMSCs during osteogenic differentiation in ONFH
RP11-216N14.9GATAD2B, ILF2, SLC27A3, CHTOP
RP11-989E6.10TP53TG3E
RP11-230G5.2HMGA2
RP11-265D17.2MICAL2, PARVA,
RP11-1060J15.4MANSC4, MRPS35, KLHL42,
RP11-794G24.1TMEM138, DDB1, CYB561A3, PGA3, TKFC,
RP11-485O10.2GALK2, COPS2, SECISBP2L
AC097532.2NCKAP5
RP11-143J24.1CHRFAM7A, GOLGA8R
RP11-762H8.3HYKK, CHRNA3, PSMA4, PSMA4, IREB2
linc-C17orf97-2DOC2B, SCGB1C2
RP11-513M1.1PIEZO2
RP11-22H5.2 CDH13
linc-ANKRD20A1-2SPATA31A3
linc-OR4M2-5GOLGA6L6
linc-ATP6V1C2-3ATP6V1C2, NOL10, PDIA6,
B, Partial lncRNAs underexpressed in hBMSCs during osteogenic differentiation in ONFH
AC104135.3TACR1
MAPT-AS1TMEM101, MPP2, CD300LG, LSM12, PYY
AL589743.1OR11H2, OR4Q3, OR4N2, OR4M1
linc-SLITRK1-4SLITRK6
AP005273.1NUDT22, GPR137, TRPT1, FKBP2, TRMT112, VEGFB
RP11-406O23.2KIAA1958, HSDL2
RP5-1102E8.3PIGK, ST6GALNAC5
linc-CSTB-3ADARB1, POFUT2,
CTD-2314B22.3OR4K1, OR4K14, OR4K2, OR4N2, OR4K15
RP5-1148A21.3EYS
LINC00473ILF2, MMP2, USP9X, CHUK, STK11, RPS6KA2
RP11-645N11.2RASA4, POLR2J3, SPDYE2,
RP11-324O2.3CCDC186, TDRD1, VWA2,

[i] hBMSCs, human bone marrow-derived mesenchymal stem cells; ONFH, osteonecrosis of the femoral head.

Bioinformatics analysis of the DNA sequence

GO analysis mainly analyzes cellular components, biological processes and molecular functions. Cellular components involved were found to include the nucleolus, cytoplasm, RISC complex, small-subunit proteasome, endomembrane system, organelle membrane, intracellular part, cytoplasmic part, membrane-bounded organelle and intracellular organelle. Biological processes involved were found to include response to oxygen radical, fatty acid beta-oxidation, protein targeting to nucleus, small molecule catabolic process, cellular biosynthetic process, protein transport, cellular lipid metabolic process, metabolic process, intracellular transport and Ras protein signal transduction. Molecular functions involved were found to include ATP binding, lipid kinase activity, chemokine receptor activity, purine ribonucleoside triphosphate binding, purine ribonucleoside binding, oxidoreductase activity, oxidizing metal ions, NAD or NADP as acceptor, ATP-dependent microtubule motor activity, plus-end-directed, transferase activity, transferring sulfur-containing groups, sulfate transmembrane transporter activity and transferase activity. The results of GO analysis are presented in Fig. 6.

Pathway analysis was conducted using the KEGG database. It was found that several pathways (Wnt, VEGF, Notch, MAPK, hedgehog, NF-κB, calcium, FoxO, PPAR and TGF-β signaling pathways, as well as mineral absorption, apoptosis and fatty acid metabolism) were involved in osteogenic differentiation in the 2 groups, as illustrated in Fig. 7.

MAPT-AS1 promotes osteogenesis and inhibits the adipo- genesis of hBMSCs

MAPT-AS1, which was downregulated in hBMSCs from patients with ONFH during osteogenic differentiation, was selected for functional analysis and for the further verification of the findings. The results revealed that the overexpression of MAPT-AS1 significantly promoted osteogenic differentiation, as indicated by ALP staining for the mineralization and expression of the osteogenic transcription factors, Runx2 and BSP (Fig. 8). On the contrary, the upregulation of MAPT-AS1 inhibited adipogenic differentiation, as indicated by Oil Red O staining and the expression of the adipogenic transcription factors, CEBP-α and PPARγ (Fig. 8).

Discussion

ONFH occurs in young individuals aged 20-40 years and 15,000-20,000 new cases of femoral head necrosis are reported annually (22,23). The causes of femoral head necrosis mainly include hormones, alcohol abuse and hip trauma. Among these, steroid-induced osteonecrosis of the femoral head (SONFH) accounts for 46.03% of total femoral head necrosis cases (23) and is the most common type of femoral head necrosis. For patients with early-stage ONFH, although early intervention can be performed through drug therapy, core decompression, interventional therapy, etc., their outcomes are not satisfactory. Approximately 65-85% of patients with femoral head necrosis will continue to develop the disease, leading to the collapse of the femoral head (24), resulting in the need for total hip replacement surgery. The majority of patients with ONFH are young adults, and the life of their prosthesis is limited; thus, they may require multiple revision surgeries in the future, which leads to a tremendous economic burden to the family and society. The aim of the present study was to explore the possible mechanisms of ONFH and provide a basis for further intervention treatment of early-stage ONFH. To the best of our knowledge, the present study is the first to describe the role of lncRNAs in hBMSCs during osteogenic differentiation in osteonecrosis of the femoral head and it has more practical clinical significance.

In recent years, scholars at home and abroad have conducted extensive research on the pathogenesis of ONFH and have proposed a multi-strand bone necrosis theory, including the intraosseous hypertension theory (25), coagulation mechanism change theory (26), lipid metabolism disorder theory (27), osteoporosis theory (28,29), bone cell apoptosis theory (30), membrane particle theory (31), gene polymorphism (32) and immune factors (33). Additionally, the disease has been found to be associated with the proliferation, osteogenic and adipogenic differentiation of hBMSCs. For example, the proliferative capability of hBMSCs has been found to be inhibited in patients with ONFH compared with healthy individuals (34). miRNA-22 has been shown to inhibit the adipogenic differentiation of hBMSCs through the protein expression of HDAC6 (35), while miRNA-100 may target BMPR2, which leads to the inhibition of osteogenic differentiation of hBMSCs (36). However, the association between lncRNAs and the osteogenic differentiation of hBMSCs during the pathogenesis of ONFH remains unclear. To date, at least to the best of our knowledge, only one study conducted focused on lncRNAs involved in femoral head necrosis (37), and studies have not been conducted on the characteristics of the osteogenic differentiation of hBMSCs from patients with ONFH.

In the present study, differentially expressed lncRNAs during the osteogenic differentiation of hBMSCs in steroid-induced femoral head necrosis were identified using the CapitalBiotech human lncRNA Array V4.0. Bioinformatics analyses, including GO and pathway analysis of differentially expressed lncRNAs, were also conducted. CNC and ceRNA networks were also analyzed. The lncRNAs identified were further verified by RT-qPCR.

The expression levels of 24 downregulated and 24 upregulated lncRNAs were determined during the osteogenic differentiation of hBMSCs in ONFH. Targets of these lncRNAs were involved in processes, such as cell proliferation, differentiation and tumor metastasis. In total, 6 lncRNAs (AC107070.1, linc-ANKRD20A1-4, RP11-794G24.1, LINC00473, MAPT-AS1 and AP005273.1) in the hBMSCs were identified and confirmed by RT-qPCR. The results of RT-qPCR revealed that the expression trends of the 4 lncRNAs were consistent with those of the microarray analysis. Moreover, hBMSCs were isolated from another 30 samples, including 15 normal and 15 patients with ONFH. The expression levels of 2 upregulated lncRNAs (AC107070.1 and linc-ANKRD20A1-4) and 2 downregulated lncRNAs (LINC00473 and MAPT-AS1) were also consistent with those of the microarray and RT-qPCR analyses, which verified the accuracy of the results.

Since the majority of the lncRNAs in current databases have not yet been functionally annotated, their functions were predicted based on the functional annotations of their related cis and trans target mRNAs. From the information presented in Table III, it was found that one lncRNA can control multiple genes, such as RP11-794G24.1, RP11-762H8.3 and AP005273.1, while one gene can be regulated by several lncRNAs. For example, OR4N2 was found to be targeted by CTD-2314B22.3 and AL589743.1. The functions of these lncRNAs were also investigated using GO analysis to determine the biological processes, cellular components and molecular functions involved. Intracellular organelle, membrane-bounded organelle and cytoplasmic part were the 3 cellular components identified. Ras protein signal transduction, intracellular transport and metabolic process were the 3 biological processes identified. Moreover, the 3 most obvious aspects of change in molecular functions were found in transferase activity, sulfate transmembrane transporter activity and transferase activity, as well as transferring sulfur-containing groups. Fatty acid metabolism, apoptosis and TGF-β signaling pathway were the 3 signaling pathways that exhibited the highest level of correlation in the KEGG pathways analysis. These 3 signaling pathways are inextricably linked to femoral head necrosis. For example, adipogenic overdifferentiation, osteoblast apoptosis and the inhibition of osteogenic differentiation through TGF-β play vital roles in the occurrence and development of femoral head necrosis. However, carbon metabolism, the pentose phosphate pathway and biosynthesis of amino acids were significantly upregulated in the KEGG analysis (37). It was hypothesized that the difference in the results was due to two aspects. First, the method used differed. In the experiments in the present study, the hBMSCs underwent osteogenic differentiation prior to microarray analysis, while in the other study, the hBMSCs were screened using microarray analysis before undergoing osteogenic differentiation. Second, differences may also be due to individual differences in the cases included.

In the present study, LINC00473, also known as LNC473, C6orf176, bA142J11.1, and is located in the 6q27 region and LINC00473, was downregulated in the hBMSCs following osteogenic differentiation. It has been found that LINC00473 was involved in the development of a number of diseases including preeclampsia (38), colorectal cancer (39), gastric cancer (40) and others. It has been demonstrated that LINC00473 is involved in the pathogenesis and development of preeclampsia and may be a candidate biomarker, as well as a therapeutic target for preeclampsia (38). Wang et al found that LINC00473 promoted Taxol resistance via miR-15a in colorectal cancer (39). Zhang and Song demonstrated that LINC00473 is an lncRNA that is associated with prognosis and malignancy in gastric cancer, while it also regulates gastric cancer cell invasion and migration (40). In the present study, the expression of LINC00473 in ONFH was found to be lower than that in normal hBMSCs, and it was hypotehsized that LINC00473 plays an important role in the osteogenic and adipogenic differentiation of stem cells via related signaling pathways.

The MAPT-AS1 gene is located in the 17q21.31 region and in the present study, MAPT-AS1 was found to be down-regulated during the osteogenic differentiation of hBMSCs. MAPT-AS1 is involved in the occurrence and development of tumors and Parkinson's disease. MAPT-AS1 overexpression has not been found in breast cancer; however, in triple-negative type (TNBC), a high MAPT-AS1 expression has been found to be associated with a longer patient survival (41). Additionally, MAPT-AS1 levels have been shown to be associated with MAPT expression, which is associated with breast cancer survival. The results of that study indicated that MAPT-AS1 may function as a potential breast cancer survival prediction biomarker (41). Pan et al (42) found that patients with ER-negative breast cancer who had larger tumors (≥2 cm), were of a younger age (<60), were at stages (III-IV) and had metastatic lymph nodes, exhibited higher levels of MAPT-AS1 expression. The regulation of natural comparable sense MAPT transcripts in cells of ER-negative breast cancer leads to the association between MAPT-AS1 and paclitaxel resistance, invasiveness and cell growth. Research has indicated that overexpression may partially protect the MAPT mRNA from degradation by the overexpression of MAPT-AS1, while the knockdown of MAPT-AS1 decreases MAPT mRNA stability. Moreover, the knockdown of MAPT also decreases MAPT-AS1 mRNA expression. MAPT-AS1 expression is coordinated with that of MAPT in breast tumor tissues (42). Moreover, MAPT-AS1 and DNMT1 have been identified as potential epigenetic regulators of MAPT expression in Parkinson's disease across 4 different brain regions and an increased MAPT expression may be associated with the disease state, but not with the neuropathology severity of Parkinson's disease (43). In the present study, the overexpres-sion of MAPT-AS1 significantly promoted the osteogenic differentiation and inhibited the adipogenic differentiation of hBMSCs at the cellular and mRNA level, as indicated by relevant staining and RT-qPCR analysis.

There were several limitations to the present study. First, the sample size of the present study was small, which may affect the results of the microarray analysis. Second, the majority of the lncRNAs require further validation by RT-qPCR. Third, the specific functions, as well as mechanisms of lncRNAs warrant further investigation.

In conclusion, to the best of our knowledge, this is the first study to elucidate the hBMSC expression profiles during osteogenic differentiation in ONFH. A total of 24 downregulated lncRNAs and 24 upregulated lncRNAs were found to be expressed during the osteogenic differentiation of hBMSCs from patients with ONFH. A bioinformatics analysis of the functions and mechanisms of the identified lncRNAs was conducted. The present study may provide a new perspective of the pathogenesis of ONFH and a novel direction for the early treatment of ONFH.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81802151), the Shandong Province Natural Science Foundation (grant nos. ZR2016HQ05, no. ZR2017BH089 and ZR2019MH012), the China Postdoctoral Science Foundation (grant no. 2018M642616) and the Qingdao Applied Foundational Research Youth Project (grant no. 19-6-2-55-cg).

Availability of data and materials

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

Authors' contributions

TL, YX, KX and YR performed the experiments and analyzed the results; YX, KX and YR wrote and drafted the manuscript; HZ and XW wrote, reviewed and edited the manuscript; HZ and YW conceived the methodology; while YJ and XW designed the research study and was a major contributor in recruiting the donors. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of the Affiliated Hospital of Qingdao University and written informed consent was obtained from all donors included in the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Guerado E and Caso E: The physiopathology of avascular necrosis of the femoral head: An update. Injury. 47(Suppl 6): S16–S26. 2016. View Article : Google Scholar

2 

Youm YS, Lee SY and Lee SH: Apoptosis in the osteonecrosis of the femoral head. Clin Orthop Surg. 2:250–225. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Calder JD, Buttery L, Revell PA, Pearse M and Polak JM: Apoptosis-a significant cause of bone cell death in osteonecrosis of the femoral head. J Bone Joint Surg Br. 86:1209–1213. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Assouline-Dayan Y, Chang C, Greenspan A, Shoenfeld Y and Gershwin ME: Pathogenesis and natural history of osteonecrosis. Semin Arthritis Rheum. 32:94–124. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Cui Q, Wang GJ, Su CC and Balian G: The otto aufranc award. Lovastatin prevents steroid induced adipogenesis and osteonecrosis. Clin Orthop Relat Res. 8–19. 1997.PubMed/NCBI

6 

Wang GJ, Sweet DE, Reger SI and Thompson RC: Fat-cell changes as a mechanism of avascular necrosis of the femoral head in cortisone-treated rabbits. J Bone Joint Surg Am. 59:729–35. 1977. View Article : Google Scholar : PubMed/NCBI

7 

Kannu P, O'Rielly DD, Hyland JC and Kokko LA: Avascular necrosis of the femoral head due to a novel C propeptide mutation in COL2A1. Am J Med Genet A. 155A:1759–1762. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S and Marshak DR: Multilineage potential of adult human mesenchymal stem cells. Science. 284:143–147. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Chen SL, Fang WW, Ye F, Liu YH, Qian J, Shan SJ, Zhang JJ, Chunhua RZ, Liao LM, Lin S and Sun JP: Effect on left ventricular function of intracoronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarction. Am J Cardiol. 94:92–95. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Wang S, Qu X and Zhao RC: Clinical applications of mesenchymal stem cells. J Hematol Oncol. 5:192012. View Article : Google Scholar : PubMed/NCBI

11 

Hare JM, Fishman JE, Gerstenblith G, DiFede VD, Zambrano JP, Suncion VY, Tracy M, Ghersin E, Johnston PV, Brinker JA, et al: Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: The POSEIDON randomized trial. JAMA. 308:2369–2379. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Liang J, Li X, Zhang H, Wang D, Feng X, Wang H, Hua B, Liu B and Sun L: Allogeneic mesenchymal stem cells transplantation in patients with refractory RA. Clin Rheumatol. 31:157–161. 2012. View Article : Google Scholar

13 

Zhao D, Cui D, Wang B, Tian F, Guo L, Yang L, Liu B and Yu X: Treatment of early stage osteonecrosis of the femoral head with autologous implantation of bone marrow-derived and cultured mesenchymal stem cells. Bone. 50:325–330. 2012. View Article : Google Scholar

14 

Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, Huarte M, Zuk O, Carey BW, Cassady JP, et al: Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature. 458:223–227. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Ponting CP, Oliver PL and Reik W: Evolution and functions of long noncoding RNAs. Cell. 136:629–641. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Guttman M, Donaghey J, Carey BW, Garber M, Grenier JK, Munson G, Young G, Lucas AB, Ach R, Bruhn L, et al: lincRNAs act in the circuitry controlling pluripotency and differentiation. Nature. 477:295–300. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Li X, Wu Z, Fu X and Han W: Long noncoding RNAs: Insights from biological features and functions to diseases. Med Res Rev. 33:517–553. 2013. View Article : Google Scholar

18 

Lee JT and Bartolomei MS: X-inactivation, imprinting, and long noncoding RNAs in health and disease. Cell. 152:1308–1323. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Wang L, Wang Y, Li Z, Li Z and Yu B: Differential expression of long noncoding ribonucleic acids during osteogenic differentiation of human bone marrow mesenchymal stem cells. Int Orthop. 39:1013–1019. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez XR, Reyes M, Lenvik T, Lund T, Blackstad M, et al: Pluripotency of mesenchymal stem cells derived from adult marrow. Nature. 418. pp. 41–49. 2002, View Article : Google Scholar

21 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

22 

Moya-Angeler J, Gianakos AL, Villa JC, Ni A and Lane JM: Current concepts on osteonecrosis of the femoral head. World J Orthop. 6:590–601. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Papakostidis C, Tosounidis TH, Jones E and Giannoudis PV: The role of 'cell therapy' in osteonecrosis of the femoral head. A systematic review of the literature and meta-analysis of 7 studies. Acta Orthop. 87:72–78. 2016. View Article : Google Scholar

24 

Pepke W, Kasten P, Beckmann NA, Janicki P and Egermann M: Core decompression and autologous bone marrow concentrate for treatment of femoral head osteonecrosis: A randomized prospective study. Orthop Rev (Pavia). 8:61622016. View Article : Google Scholar

25 

Mukisi MM, Bashoun K and Burny F: Sickle-cell hip necrosis and intraosseous pressure. Orthop Traumatol Surg Res. 95:134–138. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Kumar MN, Belehalli P and Ramachandra P: PET/CT study of temporal variations in blood flow to the femoral head following low-energy fracture of the femoral neck. Orthopedics. 37:e563–e570. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Zeng X, Zhan K, Zhang L, Zeng D, Yu W, Zhang X, Zhao M, Lai Z and Chen R: The impact of high total cholesterol and high low-density lipoprotein on avascular necrosis of the femoral head in low-energy femoral neck fractures. J Orthop Surg Res. 12:302017. View Article : Google Scholar : PubMed/NCBI

28 

Jones LC and Hungerford DS: The pathogenesis of osteonecrosis. Instr Course Lect. 56:179–196. 2007.PubMed/NCBI

29 

Kerachian MA, Séguin C and Harvey EJ: Glucocorticoids in osteonecrosis of the femoral head: A new understanding of the mechanisms of action. J Steroid Biochem Mol Biol. 114:121–128. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Jilka RL, Noble B and Weinstein RS: Osteocyte apoptosis. Bone. 54:264–271. 2013. View Article : Google Scholar :

31 

Wu Z, Ji C, Li H, Qiu G, Gao C and Weng X: Elevated level of membrane microparticles in the disease of steroid-induced vascular osteonecrosis. J Craniofac Surg. 24:1252–1256. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Zheng L, Wang W, Ni J, Li Z and Xiao T: The association of eNOS gene polymorphism with avascular necrosis of femoral head. PLoS One. 9:e875832014. View Article : Google Scholar : PubMed/NCBI

33 

Tian L, Wen Q, Dang X, You W, Fan L and Wang K: Immune response associated with Toll-like receptor 4 signaling pathway leads to steroid-induced femoral head osteonecrosis. BMC Musculoskelet Disord. 15:182014. View Article : Google Scholar : PubMed/NCBI

34 

Lee JS, Lee JS, Roh HL, Kim CH, Jung JS and Suh KT: Alterations in the differentiation ability of mesenchymal stem cells in patients with nontraumatic osteonecrosis of the femoral head: Comparative analysis according to the risk factor. J Orthop Res. 24:604–609. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Huang S, Wang S, Bian C, Yang Z, Zhou H, Zeng Y, Li H, Han Q and Zhao RC: Upregulation of miR-22 promotes osteogenic differentiation and inhibits adipogenic differentiation of human adipose tissue-derived mesenchymal stem cells by repressing HDAC6 protein expression. Stem Cells Dev. 21:2531–2540. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Zeng Y, Qu X, Li H, Huang S, Wang S, Xu Q, Lin R, Han Q, Li J and Zhao RC: MicroRNA-100 regulates osteogenic differentiation of human adipose-derived mesenchymal stem cells by targeting BMPR2. FEBS Lett. 586:2375–2381. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Wang Q, Yang Q, Chen G, Du Z, Ren M, Wang A, Zhao H, Li Z, Zhang G and Song Y: LncRNA expression profiling of BMSCs in osteonecrosis of the femoral head associated with increased adipogenic and decreased osteogenic differentiation. Sci Rep. 8:91272018. View Article : Google Scholar : PubMed/NCBI

38 

Wu D, Xu Y, Zou Y, Zuo Q, Huang S, Wang S, Lu X, He X, Wang J, Wang T and Sun L: Long noncoding RNA 00473 is involved in preeclampsia by LSD1 binding-regulated TFPI2 transcription in trophoblast cells. Mol Ther Nucleic Acids. 12:381–392. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Wang L, Zhang X, Sheng L, Qiu C and Luo R: LINC00473 promotes the Taxol resistance via miR-15a in colorectal cancer. Biosci Rep. 38:BSR201807902018. View Article : Google Scholar : PubMed/NCBI

40 

Zhang W and Song Y: LINC00473 predicts poor prognosis and regulates cell migration and invasion in gastric cancer. Biomed Pharmacother. 107:1–6. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Wang D, Li J, Cai F, Xu Z, Li L, Zhu H, Liu W, Xu Q, Cao J, Sun J and Tang J: Overexpression of MAPT-AS1 is associated with better patient survival in breast cancer. Biochem Cell Biol. 97:158–164. 2019. View Article : Google Scholar

42 

Pan Y, Pan Y, Cheng Y, Yang F, Yao Z and Wang O: Knockdown of LncRNA MAPT-AS1 inhibites proliferation and migration and sensitizes cancer cells to paclitaxel by regulating MAPT expression in ER-negative breast cancers. Cell Biosci. 8:72018. View Article : Google Scholar : PubMed/NCBI

43 

Coupland KG, Kim WS, Halliday GM, Hallupp M, Dobson-Stone C and Kwok JB: Role of the long non-coding RNA MAPT-AS1 in regulation of microtubule associated protein Tau (MAPT) expression in Parkinson's disease. PLoS One. 11:e01579242016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2020
Volume 46 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li T, Xiao K, Xu Y, Ren Y, Wang Y, Zhang H, Weng X and Jiang Y: Identification of long non‑coding RNAs expressed during the osteogenic differentiation of human bone marrow‑derived mesenchymal stem cells obtained from patients with ONFH. Int J Mol Med 46: 1721-1732, 2020
APA
Li, T., Xiao, K., Xu, Y., Ren, Y., Wang, Y., Zhang, H. ... Jiang, Y. (2020). Identification of long non‑coding RNAs expressed during the osteogenic differentiation of human bone marrow‑derived mesenchymal stem cells obtained from patients with ONFH. International Journal of Molecular Medicine, 46, 1721-1732. https://doi.org/10.3892/ijmm.2020.4717
MLA
Li, T., Xiao, K., Xu, Y., Ren, Y., Wang, Y., Zhang, H., Weng, X., Jiang, Y."Identification of long non‑coding RNAs expressed during the osteogenic differentiation of human bone marrow‑derived mesenchymal stem cells obtained from patients with ONFH". International Journal of Molecular Medicine 46.5 (2020): 1721-1732.
Chicago
Li, T., Xiao, K., Xu, Y., Ren, Y., Wang, Y., Zhang, H., Weng, X., Jiang, Y."Identification of long non‑coding RNAs expressed during the osteogenic differentiation of human bone marrow‑derived mesenchymal stem cells obtained from patients with ONFH". International Journal of Molecular Medicine 46, no. 5 (2020): 1721-1732. https://doi.org/10.3892/ijmm.2020.4717