Skip to main content
Erschienen in: Arthritis Research & Therapy 1/2018

Open Access 01.12.2018 | Research article

Aberrantly expressed messenger RNAs and long noncoding RNAs in degenerative nucleus pulposus cells co-cultured with adipose-derived mesenchymal stem cells

verfasst von: Zhihua Han, Jiandong Wang, Liang Gao, Qiugen Wang, Jianhong Wu

Erschienen in: Arthritis Research & Therapy | Ausgabe 1/2018

Abstract

Background

Stem cell therapy is considered as a promising alternative to treat intervertebral disc degeneration (IDD). Extensive work had been done on identifying and comparing different types of candidate stem cells, both in vivo and in vitro. However, few studies have shed light on degenerative nucleus pulposus cells (NPCs), especially their biological behavior under the influence of exogenous stem cells, specifically the gene expression and regulation pattern. In the present study, we aimed to determine messenger RNAs (mRNAs) and long non-coding RNAs (lncRNAs), which are differentially expressed during the co-culturing process with adipose-derived mesenchymal stem cells (ASCs) and to explore the involved signaling pathways and the regulatory networks.

Methods

We compared degenerative NPCs co-cultured with ASCs with those cultured solely using lncRNA-mRNA microarray analysis. Based on these data, we investigated the significantly regulated signaling pathways based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database. Moreover, 23 micro RNAs (miRNAs), which were demonstrated to be involved in IDD were chosen; we investigated their theoretic regulatory importance associated with our microarray data.

Results

We found 632 lncRNAs and 1682 mRNAs were differentially expressed out of a total of 40,716 probes. We then confirmed the microarray data by real-time PCR. Furthermore, we demonstrated 197 upregulated, and 373 downregulated Gene Ontology terms and 176 significantly enriched pathways, such as the mitogen-activated protein kinase (MAPK) pathway. Also, a signal-net was constructed to reveal the interplay among differentially expressed genes. Meanwhile, a mRNA-lncRNA co-expression network was constructed for the significantly changed mRNAs and lncRNAs. Also, the competing endogenous RNA (ceRNA) network was built.

Conclusion

Our results present the first comprehensive identification of differentially expressed lncRNAs and mRNAs of degenerative NPCs, altered by co-culturing with ASCs, and outline the gene expression regulation pattern. These may provide valuable information for better understanding of stem cell therapy and potential candidate biomarkers for IDD treatment.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13075-018-1677-x) contains supplementary material, which is available to authorized users.
Zhihua Han and Jiandong Wang contributed equally to this work.
Abkürzungen
ACAN
Aggrecan
ASCs
Adipose-derived mesenchymal stem cells
ceRNA
Competing endogenous RNA
COL2A1
Type II collagen
ENPP1
Ectonucleotide pyrophosphatase 1
FDR
False discovery rate
GEO
Gene Expression Omnibus
GO
Gene Ontology
GSK3B
Glycogen synthase kinase 3 beta
IDD
Intervertebral disc degeneration
ITGA
Integrin alpha
KEGG
Kyoto Encyclopedia of Genes and Genomes
lncRNAs
Long non-coding RNAs
MAPK
Mitogen-activated protein kinase
miRNAs
Micro RNAs
MREs
Micro RNA response elements
mRNA
Messenger RNA
MSCs
Mesenchymal stem cells
MT1F
Metallothionein 1F
NF-kappa B
Kappa-light-chain-enhancer of activated B cells
NP
Nucleus pulposus
NPCs
Nucleus pulposus cells
OPN
Osteopontin
PI3K
Phosphoinositide 3-kinase
PIK3CB
Phosphoinositide-3-kinase, catalytic, beta polypeptide
PIK3R3
Phosphoinositide-3-kinase, regulatory subunit 3
Pol II
Polymerase II
RVM
Random variance model
SOX9
Sex determining region Y box 9
SPP1
Secreted phosphoprotein 1
XIST
X-inactive specific transcript

Background

Intervertebral disc degeneration (IDD) due to aging and other reasons is still a major clinical challenge for spine surgeons. Numerous strategies have been studied to prevent the deterioration and progressing of IDD, which will cause various pathologies, such as disc herniation, spinal stenosis and segmental instability [1]. However, disc degeneration is difficult to reverse in the natural course due to the avascular and aneural nature of the nucleus pulposus (NP). Biochemically, the degeneration is characterized by the decrease of the extracellular matrix and loss of water content within the NP, which are believed to play a crucial role in the progress of IDD.
Recently, stem cell therapy, introducing mesenchymal stem cells (MSCs) or cytokines into target discs, appears as a promising strategy for the IDD treatment. This emerging modality was proved to be effective both in vitro [24] and in vivo [5, 6]. Co-culturing with nucleus pulposus cells (NPCs), MSCs could differentiate towards the NP-like cells with increased expression of the NP marker genes [7]. In particular, adipose-derived mesenchymal stem cells (ASCs) co-cultured with NPCs expressed significantly upregulated expression of NP-related genes including sex determining region Y box 9 (SOX9), type II collagen (COL2A1), and aggrecan (ACAN) [8]. Interestingly, our previous study demonstrated that degenerative NPCs could be activated by MSCs in the co-culture system with significantly upregulated gene expression of SOX9, COL2A1, and ACAN [4]. It is notable that degenerative NPCs could be induced towards healthy NPCs by specific cytokines and cellular factors yielded from MSCs [9]. Also, it underlines that the gene expression pattern of not only MSCs but also degenerative NPCs could be altered through the intracellular cross-talking [10, 11]. However, the underlying mechanism of these phenomena is still not fully understood.
Gene microarray technology can simultaneously measure differences in the expression level of thousands of genes of predefined groups of samples [12] and allows highly effective evaluation of genome-wide expression changes [13]. This novel technology enables researchers to develop a more comprehensive understanding of the cross-talking between MSCs and degenerative NPCs, including differentially expressed genes and long non-coding RNAs (lncRNAs).
Recently, lncRNAs have received critical attention with their regulatory effect on gene expression [14, 15]. They have been characterized with the high tissue-specificity and low sequence-conservation [16, 17] and have been demonstrated to be involved in various physiological and pathological processes as regulators, such as imprinting, X-inactivation, and development [18]. Numerous previous studies have attempted to map the phenotype of NPCs [1922], to the best of our knowledge, yet few studies have emphasized the function of lncRNAs related to disc degeneration, particularly their regulatory roles in the process of degenerative NPCs co-cultured with MSCs [23].
Similarly, micro RNAs (miRNAs), a group of small and non-coding RNAs, have been proved to participate in the expression regulation of coding genes and to influence various biological processes, including cell differentiation, proliferation, and metabolism [15, 24]. Many miRNAs have been demonstrated to be involved in natural disc degeneration [25, 26], and specifically miR-27b [27] and miR-93 [28] have been shown to promote matrix degradation within the discs and accelerate the disc degeneration process. Yet, there was no study focusing on the role of these miRNAs in the cell-cell cross-talk between MSCs and degenerative NPCs.
The current study aimed to use gene expression microarray analysis and bioinformatics methods to investigate the effect of MSCs on degenerative NPCs in terms of deferentially expressed lncRNAs and mRNAs, signaling pathways, and gene regulation networks involving mRNAs, lncRNAs, and miRNAs (Additional file 1).

Methods

All human tissues were obtained and used with informed consent from the patients and under the approval of the Institutional Review Broad of the Shanghai General Hospital, Shanghai Jiaotong University.

Isolation and culture of NP cells

The human NP tissue was surgically obtained from the degenerative discs (grade III–IV according to the Pfirrmann grading system [29]) of three patients diagnosed as having lumbar spondylosis. The isolation and culture of NPCs was performed as previously reported [4, 30] (Additional file 2).

Isolation and culture of ASCs

The adipose tissue surgically obtained from the patients’ backs was processed for isolation of ASCs following the previously standardized protocol [31, 32] (Additional file 2).

Co-culture of ASCs and NPCs

Both NPCs and ASCs at passage 3 were co-cultured using the non-direct cell-cell contact co-culturing system, consisting of six-well plates and polyethylene terephthalate track-etched tissue culture inserts with 0.4-μm pore size. Briefly, ASCs (6.0 × 104 cells) were seeded on the base of the six-well plate, and the same numbers of NPCs were seeded onto the upper surface of the membrane. Co-cultured cells were maintained for 7 days at 37 °C and 5% CO2 in a humidified atmosphere with the medium being changed every 2 days. Meanwhile, the NPCs (6.0 × 104 cells) at passage 3 were cultured for 7 days in the same condition as the control.

RNA isolation and quality control

Briefly, the total RNA was isolated from each group of cells using the Trizol agent (Invitrogen, Carlsbad, USA) following the manufacture’s protocol. Then the RNA was purified with an RNase Kit (Bio-Rad, CA, USA), and the quantity was measured using a spectrophotometer (NanoDrop-1000, Thermo Scientific, MA, USA). Agarose-gel electrophoresis was performed to test the RNA integrity and DNA contamination (Additional file 3).

Microarray analysis

Generally, NPCs without co-culture (control group) and with co-culture (experimental group) were used to compare mRNA and lncRNA expression profiles. As shown in additional file 1, a multiple-step strategy was used to identify mRNAs and lncRNAs dysregulated between NPCs with and without co-culturing. To clarify the changes in the signaling pathways of NPCs during co-culture, we further performed Gene Ontology (GO) analysis, pathway analysis, and signal-net analysis. Microarray analysis was performed by the GMINIX Informatics Ltd. Co (Shanghai, China). The quality control of hybridization is shown in Additional file 4. The data had been uploaded to the NCBI Gene Expression Omnibus (GEO) and can be accessed [GEO:GSE112216] (https://​www.​ncbi.​nlm.​nih.​gov/​geo/​query/​acc.​cgi?​acc=​GSE112216).
The mRNAs and lncRNAs differently expressed between NPCs with and without co-culturing were identified using the random variance model (RVM) t test. The RVM t test was applied to filter the differentially expressed RNAs with increased degrees of freedom in the small sample datasets. With a threshold of P < 0.05 considered significant, differentially expressed RNAs were selected and false discovery rate (FDR) analysis was performed. Unsupervised hierarchical clustering was performed and a cluster map was created.

Real-time PCR

To validate the microarray results, seven mRNAs were selected for the real-time PCR validation. Complementary DNA (cDNA) was generated by the reverse transcript using a Taqman Reverse Transcription Kit (Invitrogen, Carlsbad, USA) according to the manufacturer’s instructions. Gene expression analysis was conducted by real-time PCR using the SYBR Green Mastermix (BioRad, CA, USA) and a CFX96 Touch Real-time PCR Detection System (BioRad, CA, USA). Homo actin was used as the internal control to determine the relative expression of target genes; the relative changes in gene expression were compared to those of untreated cells using the 2-ΔΔCT method where CT = threshold cycle. All reactions were performed in triplicate and the sequences of used primers are shown in Table 1.
Table 1
Primers used in real-time PCR
Gene name
Forward sequence 5′-3′
Reverse sequence 5′-3’
PIK3R3
GGG GAA GTG AAG CAC TGT GT
GAC GTT GAG GGA GTC GTT GT
ENPP1
GCC CGA AAT CTT TCT TGC CG
TGC CAT GCT TGA ATC CAG GT
MT1F
TGC AAG TGC AAA GAG TGC AA
CCC TTT GCA AAC ACA GCC C
SPP1
GCC GAG GTG ATA GTG TGG TT
AAC GGG GAT GGC CTT GTA TG
EPYC
TTC TGG GGC CAC ACA CAA AT
GCT CTC GAA GTT GAG GCA GT
CD24
GCT CCT ACC CAC GCA GAT TT
GAG ACC ACG AAG AGA CTG GC
C4orf31
TCA TGT CTA CTC CAG GCC CA
GTA GTA CTG CGT GTC GGG TT
homo-actin
GCT CAG GAG GAG CAA TGA TCT TG
GTA CGC CAA CAC AGT GCT GTC
PIK3R3 phosphoinositide-3-kinase, regulatory subunit 3, ENPP1 ectonucleotide pyrophosphatase/ phosphodiesterase 1, MT1F metallothionein 1F, SPP1 secreted phosphoprotein 1, EPYC epiphycan, C4orf31 chromosome 4 open reading frame 31

GO analysis

Based on the GO database (http://​www.​geneontology.​org), the GO analysis was performed to analyze the main functions of the differentially expressed mRNAs using the two-sided Fisher’s exact test and chi-square test. The differentially expressed genes were evaluated independently and classified to upregulation and downregulation. P values of all differentially expressed genes were computed in all GO categories, and P < 0.01 was defined as significant.

Pathway analysis

The significance levels of pathways associated with differentially expressed genes were analyzed based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (http://​www.​genome.​jp/​kegg/​). Fisher’s exact test and the chi-square test were used to select the significant pathways according to the significance threshold P < 0.05.
In order to systematically identify the integrations between the pathways, the Path-net, highlighting the interaction net containing the pathways associated with differentially expressed genes, was generated based on the interactions among the pathways of the KEGG database.

Signal-net analysis

The significant intersectional genes in both GO analysis and Pathway analysis were selected to analyze the gene-gene interaction and construct the network map. From the differentially expressed gene data, the gene-gene network map was constructed based on the KEGG database, allowing the users to build and analyze the molecular networks. The networks are stored and presented as graphs, the nodes are genes, and edges representing relationship types between the nodes may indicate activation or phosphorylation. The graph nature of networks allowed further investigation with the powerful tools implemented in R. The network work of each gene was calculated by counting the numbers of upstream genes and downstream genes, which were expressed in the form of in-degree and out-degree. The betweenness centrality of each gene was calculated according to its in-degree and out-degree, and higher betweenness centrality implies greater importance in the gene-network regulation.

LncRNA-gene-net analysis

Co-expression network analysis of lncRNAs and mRNAs was performed based on the differentially expressed lncRNAs and the intersectional mRNAs, which were significant in both GO analysis and Pathway analysis.

Competing endogenous RNA (ceRNA) analysis

The miRNAs are a class of ~ 22-nucleotide-long single-stranded non-coding RNAs that regulate gene expression by binding to miRNA response elements (MREs) on the RNAs [33, 34]. The lncRNAs are non-coding RNAs longer than 200 nucleotides and also involved in the pathology of many complex human diseases including cancer [35]. The lncRNAs also harbor MREs and compete with other RNAs for the miRNA binding, and lncRNAs can regulate miRNA abundance by sequestering and binding them [36], thus functioning to compete with endogenous RNAs to influence post-transcriptional regulation.
Based on previous studies, 23 miRNAs associated with disc degeneration (Additional file 5) were selected to investigate their regulatory involvement in differentially expressed mRNAs and lncRNAs defined by our study. Then the miRNA-mRNA target prediction according to TargetScan (http://​www.​targetscan.​org/) and the miRanda (http://​cbio.​mskcc.​org/​miRNA2003/​miranda.​html) was performed for competing endogenous RNA. The ceRNA network was thereafter constructed based on those negatively regulated intersectional lncRNAs and mRNAs.

Statistical analysis

All data are reported as mean ± standard deviation. Differences between groups were evaluated by Student’s t test using SPSS 20.0 software (Chicago IL, USA.). P < 0.05 was considered statistically significant.

Results

Validation of ASCs

To verify the ASCs, specific surface markers and multiple differentiation potentials were verified. On flow cytometry, positive expression of CD90, CD105, and HLA-ABC and negative expression of CD34, CD45, and HLA-DR was observed (Fig. 1). Also, the multilineage differentiation potential of ASCs was proved by the histological staining (Fig. 1).

Identification of deferentially expressed lncRNAs and mRNAs

A total of 2314 probes were identified to be differentially expressed, including 632 lncRNAs and 1682 mRNAs (Fig. 2a). For lncRNAs, the chromosome-3 open reading frame 49 (C3orf49) showed the greatest upregulation (fold change = 18.9), followed by the two-pore channel 3 pseudogene, upregulated (LOC440895) and testis-specific transcript Y-linked 15, upregulated (TTTY15). Additionally, the top three deferentially expressed mRNAs were the secreted phosphoprotein 1 (SPP1), metallothionein 1F (MT1F) and ectonucleotide pyrophosphatase 1 (ENPP1) with their fold changes were 106, 77, and 34, respectively (Fig. 2b). Furthermore, these deferentially expressed mRNAs also include NPCs maker mRNAs such as SOX-9 and COL2A1. More detailed information is provided in Additional file 6 (top 10 differentially expressed lncRNAs and mRNAs) and Additional file 7 (full list of differentially expressed mRNAs and lncRNAs).

Validation of real-time PCR

Furthermore, we demonstrated that the results of the real-time PCR analysis were consistent with the microarray data. Seven differentially expressed mRNAs (PIK3R3, ENPP1, MT1F, SPP1, EPYC, CD24, and C4orf31) were selected for the real-time PCR analysis. The gene chip analysis revealed these mRNAs were upregulated up to 2.70-fold, 34.48-fold, 76.92-fold, 106.38-fold, 34.58-fold, 20.40-fold, and 17.85-fold, respectively. The microarray analysis data were also verified, and the expression of PIK3R3 (P < 0.05), ENPP1 (P < 0.05), MT1F (P < 0.05), SPP1 (P < 0.05), EPYC (P < 0.05), CD24 (P < 0.05), C4orf31 (P < 0.05) in the NPCs were significantly increased up to 11.32-fold, 70.92-fold, 192.01-fold, 1896.14-fold, 1132.71-fold, 31.87-fold, and 94.45-fold, respectively (Fig. 3).

GO analysis

Differentially expressed mRNAs and lncRNAs were used for the downstream GO analysis and pathway analysis. There were 197 upregulated and 373 downregulated GO terms (P < 0.05), including the upregulated cell adhesion (GO:0007155) and the downregulated small molecule metabolic process (GO:0044281). The top 10 upregulated and downregulated GO terms are presented in Fig. 4.

Pathway analysis

The pathway analysis identified a total of 176 pathways, among which 122 pathways were statistically significantly upregulated and 54 were significantly downregulated. The most enriched pathways included phosphoinositide 3-kinase (PI3K)-protein kinase PI3K-Akt signaling pathway (upregulated) and metabolic pathways (downregulated) (Fig. 5).
The interaction network for all significantly enriched pathways is formulated with the Path-net (Fig. 6; Additional file 8), representing these pathways directly and systematically involved in the interaction between ASCs and NPCs. Also, we discovered that the mitogen-activated protein kinase (MAPK) signaling pathway played a canonical role with the highest number of interactions with other pathways (interaction degree, 35) (Fig. 6). In contrast, the apoptosis pathway (interaction degree, 25) and the Wnt signaling pathway (interaction degree, 18) were downregulated in the co-culture system.

Signal-net

Based on the intersectional genes that were significantly enriched in both GO analysis and Pathway enrichment analysis, a gene interaction network was constructed (Fig. 7; Additional file 9). Signal-net analysis showed that phosphoinositide-3-kinase, regulatory subunit 3, upregulated (PIK3R3), phosphoinositide-3-kinase, catalytic, beta polypeptide, downregulated (PIK3CB) and fibroblast growth factor receptor 2, downregulated (FGFR2) are critical in the gene regulatory network with their degrees of connections 19, 18 and 15, respectively.

lncRNA-mRNA co-expression network

A co-expression network was built for 80 lncRNAs and 170 mRNAs selected from the differentially expressed lncRNAs and mRNAs based on the degree of correlation (Fig. 8). These 250 RNAs (250 nodes in the co-expression network) were further combined into 1453 pairs of co-expression lncRNA-mRNA. Among these RNAs of the co-expression network, X-inactive specific transcript, (non-coding, downregulated, degree, 77 (XIST) obtained the highest number of interactions followed by the TCONS_l2_00013892-XLOC_l2_007489 (non-coding, upregulated, degree, 69) and TCONS_00020478-XLOC_009810 (non-coding, upregulated, degree, 66).

Regulatory role of miRNAs

A competing endogenous RNA network was constructed from 23 miRNAs, previously proved to be involved in intervertebral disc degeneration, and displayed their regulatory interplay with differentially expressed mRNAs and lncRNAs (Fig. 9, Additional file 5). Evidently, hsa-miR-98-5p (upregulated, interaction degree, 57) was the most important miRNA in this network, followed by the hsa-miR-27a-3p (upregulated, interaction degree, 40) and hsa-miR-146a-3p (upregulated, interaction degree, 23). Additionally, TCONS_l2_00011557-XLOC-_l2_005806 TCONS_l2_00010454-XLOC_l2_005438, and TCONS_l2_00023712-XLOC_l2_011987 were the top three important lncRNAs in this regulatory network; their interaction degrees were 53, 44, and 35, respectively (Additional file 6 and Additional file 7).

Discussion

In the present study, we identified a significant number of mRNAs and lncRNAs differentially expressed by degenerative NPCs co-cultured with ASCs, using the microarray analysis and in-depth data profiling. We also identified the signaling pathways that were altered during the co-culture and outlined the co-expression relationship between mRNAs and lncRNAs. The disc-degeneration-related miRNAs, differentially expressed mRNAs, and lncRNAs were further evaluated to identify the regulatory interaction, highlighting the biological pathways and cellular events of gene expression and regulation during the stem-cell therapy process. In addition, our research validates the previous studies about NPC phenotypes [1922] and further investigated the regulatory role of lncRNAs. However, more in-depth understanding of these gene expression and regulation profiles will provide valuable clues for gene therapy approaches for disc degeneration.
In the present study, we revealed the altered mRNAs and lncRNAs between NPCs before and after co-culture with ASCs. Real-time PCR results of seven randomly selected mRNAs (Fig. 3) were consistent with the microarray data, further confirming the high credibility of the microarray analysis. Furthermore, we identified that the NP marker gene expression, SOX-9 (fold change = 7, P < 0.001) and COL2A1 (fold change = 6, P < 0.001), was increased in NPCs after the co-culture, indicating the upregulated synthesis and secretory activities of NPCs under the influence of MSCs [37, 38]. These findings were consistent with the previous studies reporting the significantly raised expression of SOX-9 and COL2A1 in NPCs when co-cultured with MSCs [39, 40].
In particular, we demonstrated that SPP1 was the most significantly altered gene and might be indicated as a marker of NPC (Additional file 6). SPP1, also known as osteopontin (OPN), is an extracellular structural protein secreted by various types of cells. Our data from the signal-net analysis suggested that SPP1 could activate the integrin proteins, e.g. integrin alpha (ITGA) 1, 3, 4, 6, 7, 8, and 10, and has close connections with COL11A1, COL3A1, and COL2A1 (Fig. 7). Since integrins are a class of cell adhesion molecules that regulate interactions between a cell and its surrounding matrix [41], activation of ITGA will assist NPCs to interact with the extracellular matrix, specifically collagen molecules, and then to potentially restore the intervertebral disc function. Marfia and colleagues previously reported greater expression of SPP1/OPN and CD44 in degenerative IVD comparing to herniated IVD, and SPP1/OPN was only detected in degenerative IVD tissue [42]. They therefore assumed that SPP1/OPN might mark the severity of disc degeneration. However, in disagreement with Marfia’s study, our data revealed that SPP1/OPN was upregulated with downregulated CD44 when degenerative NPCs were co-cultured with ASCs (Additional file 7). Such a considerable divergence warrants further in-depth investigations into the sophisticated underlying mechanism.
We also demonstrated that CD24 were upregulated (Additional file 6) and these data were in accord with previous studies, signifying CD24 as an important marker of NPCs and notochordal cells [22, 4348]. Particularly, Ricardo Rodrigues-Pinto et al. demonstrated that CD24, a glycosylphosphatidylinositol anchor protein, is one of the notochord-specific markers during the early development of human IVD [47]. Similarly, Nobuyuki Fujita et al. [46] identified CD24 as a surface marker for NPC with its high expression in the healthy and herniated NP tissue rather than in the annulus fibrosus. Furthermore, CD24 is proved to be a key marker of the irreversible cellular hierarchy during the differentiation process of the NP-progenitor cells towards NP-committed cells in mice and humans [49]. Therefore, in our study, the upregulated expression of CD24 also confirmed the positive effect of ASCs on NPC regeneration.
Functional annotation of these differentially expressed mRNAs and lncRNAs was investigated by GO and KEGG pathway analysis. The GO analysis identified significant enrichments in over 197 GO terms, including cell adhesion (GO:0007155), positive regulation of transcription from RNA polymerase II (Pol II) promoter (GO:0045944), and extracellular matrix organization (GO:0030198). These data indicate that the metabolic activities of NPCs might be enhanced by ASCs. Specifically, the cellular adhesion is crucial for stable connections between cells and tissue structure maintenance, also involved in diverse signal transduction [50]. The upregulated RNA Pol II promoter of co-cultured NPCs in the present study reflects the increased expression of protein-encoding genes. The RNA Pol II together with other factors, mediating the transcription initiation of protein-encoding genes, is an essential control point for gene expression in the eukaryotes [51]. Similarly, upregulated extracellular matrix organization would counteract the loss of extracellular matrix and further favor the regeneration of NP tissue [52].
Additionally, the most important signaling pathways altered during the co-culture were the MAPK pathway, the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kappa B) signaling pathway, and the PI3K-Akt signaling pathway (Fig. 5). Specifically, the NF-kappa B and MAPK signaling pathways, the principal regulators of inflammation and catabolism [53], are important in the symptomatic disc-degenerative diseases [5456]. The MAPK signaling pathway was generally increased in the aged and degenerative discs [57, 58]. Our analysis displayed the downregulation of the MAPK signaling pathway, indicating degenerative NPCs could be stimulated towards normal NPCs by ASCs. In addition, NF-kappa B targets several pro-inflammatory cytokines [5961] that are highly expressed in degenerative discs rather than normal discs. Therefore, the downregulated NF-kappa B signaling pathway of co-cultured NPCs in our study may indicate that ASCs protect NPCs from inflammatory response. Furthermore, the PI3K-Akt signaling pathway was identified as the most significantly upregulated pathway. Activated PI3K-Akt can protect against the disc degeneration [62, 63] with increased extracellular matrix synthesis [64], promoted cell proliferation [65], counteraction of cell apoptosis [66], and alleviated oxidative damage [67].
Moreover, the signal-net analysis displayed that glycogen synthase kinase 3 beta (GSK3B), which was downregulated, plays the most critical role in the network with the highest betweenness centrality. As a serine/threonine kinase, GSK3 is involved in the phosphorylation of numerous substrates, including signaling proteins, transcription factors and structural proteins [68, 69]. As a crucial mediator of PI3K-Akt, PKA, PKC, and Wnt/ß-catenin, GSK3 is proved to play an important role in chondrocyte differentiation [7072]. Miclea et al. demonstrated that GSK3B could inhibit the chondrocyte proliferation and increase the cartilage apoptosis via activating the canonical Wnt signaling pathway in the ex vivo mouse embryos [73]. Also, Itoh et al. demonstrated that GSK3 proteins are involved in early stages of chondrocyte differentiation by driving the differentiation in a cell-autonomous manner [74]. In the present study, we also found that after co-culture, the degenerative NPCs expressed significantly higher SOX9 and COL1A2, confirming the restored function of the NPCs.
Regarding the regulatory functions of lncRNAs, in the present study we investigated the co-expressional connection between mRNAs and lncRNAs and found that XIST acquired the greatest number of interactions (degree = 77) among all RNAs. XIST, a 17–20 kb RNA, binds the X chromosome [75, 76] to initiate X chromosome inactivation [77] and is required for whole-chromosome silencing [78]. Also, XIST provides one of a few tangible readouts for the stem cell quality [79] and also influences the pluripotent stem cell population, as proved in induced pluripotent stem cell treatments in regenerative medicine [80]. However, such significant roles have not studied in the context of disc degeneration and further investigations are warranted.
A competing endogenous RNAs network was constructed for the regulatory function of various lncRNAs and many miRNAs associated with disc degeneration. miR-98-5p is the most important in the network (interaction degree 57) followed by the miR-27a-34 (interaction degree, 40) (Fig. 9). miR-98 is significantly downregulated in the degenerated NP tissue and has been proved to promote type II collagen expression in NPCs [81]. Also, Li et al. reported that the downregulation of miRNA-27b would yield loss of type II collagen and lead to the development of IDD [27]. Therefore, the upregulation of these two miRNAs, favoring type II collagen synthesis of the NPCs, demonstrated the positive effect of ASCs on degenerative NPCs and might serve as potential therapeutic targets in IDD.
This study has some limitations. First, numerous RNA probes were investigated in the microarray analysis and this limited the validation of the gene chip results. Therefore, we only interpreted the results based on previous studies and our interests. Also, to avoid losing information, genes and lncRNAs were not further classified into specific subsets according to their functions and only the regulatory roles of differentially expressed RNAs of general interest were interpreted, serving as potential targets of the further in-depth investigation. Nevertheless, the results from our bioinformatics analysis only elucidate relevant relationships associated with these genes and lncRNAs; more in vitro or in vivo studies are necessary to comprehensively understand the specific involvement of the differentially expressed lncRNAs and mRNAs during the co-culturing process.

Conclusion

To sum up, co-culturing human ASCs with degenerative NPCs restored the biological status of the degenerative NPCs. Our study identified the interplay between ASCs and degenerative NPCs during the co-culturing and provided valuable information for the development and application of gene therapy for IDD. More studies are required to explore the functions and mechanisms of the key RNAs involved in the regeneration of the human intervertebral disc tissue and further benefit the translation of gene therapy in IDD from bench to bedside.

Funding

This study was supported by the National Natural Science Foundation of China (grant number 81301580).

Availability of data and materials

The gene microarray datasets had been uploaded to the NCBI Gene Expression Omnibus (GEO) and can be accessed via GEO Series accession [GEO:GSE112216] (https://​www.​ncbi.​nlm.​nih.​gov/​geo/​query/​acc.​cgi?​acc=​GSE112216). Other related data generated and/or analyzed during the current study are available from the corresponding author upon reasonable request.
All human tissues were obtained and used with the informed consent of the patients and under the approval of Institutional Review Broad of the Shanghai General Hospital, Shanghai Jiaotong University.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Cao C, Zou J, Liu X, Shapiro A, Moral M, Luo Z, et al. Bone marrow mesenchymal stem cells slow intervertebral disc degeneration through the NF-kappaB pathway. Spine J. 2015;15:530–8.CrossRefPubMed Cao C, Zou J, Liu X, Shapiro A, Moral M, Luo Z, et al. Bone marrow mesenchymal stem cells slow intervertebral disc degeneration through the NF-kappaB pathway. Spine J. 2015;15:530–8.CrossRefPubMed
2.
Zurück zum Zitat Vadala G, Studer RK, Sowa G, Spiezia F, Lucu C, Denaro V, et al. Coculture of bone marrow mesenchymal stem cells and nucleus pulposus cells modulate gene expression profile without cell fusion. Spine (Phila Pa 1976). 2008;33:870–6.CrossRef Vadala G, Studer RK, Sowa G, Spiezia F, Lucu C, Denaro V, et al. Coculture of bone marrow mesenchymal stem cells and nucleus pulposus cells modulate gene expression profile without cell fusion. Spine (Phila Pa 1976). 2008;33:870–6.CrossRef
3.
Zurück zum Zitat Watanabe T, Sakai D, Yamamoto Y, Iwashina T, Serigano K, Tamura F, et al. Human nucleus pulposus cells significantly enhanced biological properties in a coculture system with direct cell-to-cell contact with autologous mesenchymal stem cells. J Orthop Res. 2010;28:623–30.PubMed Watanabe T, Sakai D, Yamamoto Y, Iwashina T, Serigano K, Tamura F, et al. Human nucleus pulposus cells significantly enhanced biological properties in a coculture system with direct cell-to-cell contact with autologous mesenchymal stem cells. J Orthop Res. 2010;28:623–30.PubMed
4.
Zurück zum Zitat Han Z, Zhang Y, Gao L, Jiang S, Ruan D. Human Wharton’s jelly cells activate degenerative nucleus pulposus cells in vitro. Tissue Eng Part A. 2018;24:1035–43. Han Z, Zhang Y, Gao L, Jiang S, Ruan D. Human Wharton’s jelly cells activate degenerative nucleus pulposus cells in vitro. Tissue Eng Part A. 2018;24:1035–43.
5.
Zurück zum Zitat Zhang Y, Tao H, Gu T, Zhou M, Jia Z, Jiang G, et al. The effects of human Wharton’s jelly cell transplantation on the intervertebral disc in a canine disc degeneration model. Stem Cell Res Ther. 2015;6:154.CrossRefPubMedPubMedCentral Zhang Y, Tao H, Gu T, Zhou M, Jia Z, Jiang G, et al. The effects of human Wharton’s jelly cell transplantation on the intervertebral disc in a canine disc degeneration model. Stem Cell Res Ther. 2015;6:154.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Chun HJ, Kim YS, Kim BK, Kim EH, Kim JH, Do BR, et al. Transplantation of human adipose-derived stem cells in a rabbit model of traumatic degeneration of lumbar discs. World Neurosurg. 2012;78:364–71.CrossRefPubMed Chun HJ, Kim YS, Kim BK, Kim EH, Kim JH, Do BR, et al. Transplantation of human adipose-derived stem cells in a rabbit model of traumatic degeneration of lumbar discs. World Neurosurg. 2012;78:364–71.CrossRefPubMed
7.
Zurück zum Zitat Vadala G, Russo F, Ambrosio L, Loppini M, Denaro V. Stem cells sources for intervertebral disc regeneration. World J Stem Cells. 2016;8:185–201.CrossRefPubMedPubMedCentral Vadala G, Russo F, Ambrosio L, Loppini M, Denaro V. Stem cells sources for intervertebral disc regeneration. World J Stem Cells. 2016;8:185–201.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Sun Z, Liu ZH, Zhao XH, Sun L, Chen YF, Zhang WL, et al. Impact of direct cell co-cultures on human adipose-derived stromal cells and nucleus pulposus cells. J Orthop Res. 2013;31:1804–13.PubMed Sun Z, Liu ZH, Zhao XH, Sun L, Chen YF, Zhang WL, et al. Impact of direct cell co-cultures on human adipose-derived stromal cells and nucleus pulposus cells. J Orthop Res. 2013;31:1804–13.PubMed
9.
Zurück zum Zitat Smith LJ, Nerurkar NL, Choi KS, Harfe BD, Elliott DM. Degeneration and regeneration of the intervertebral disc: lessons from development. Dis Model Mech. 2011;4:31–41.CrossRefPubMed Smith LJ, Nerurkar NL, Choi KS, Harfe BD, Elliott DM. Degeneration and regeneration of the intervertebral disc: lessons from development. Dis Model Mech. 2011;4:31–41.CrossRefPubMed
10.
Zurück zum Zitat Lehmann TP, Filipiak K, Juzwa W, Sujka-Kordowska P, Jagodzinski PP, Zabel M, et al. Coculture of human nucleus pulposus cells with multipotent mesenchymal stromal cells from human bone marrow reveals formation of tunnelling nanotubes. Mol Med Rep. 2014;9:574–82.CrossRefPubMed Lehmann TP, Filipiak K, Juzwa W, Sujka-Kordowska P, Jagodzinski PP, Zabel M, et al. Coculture of human nucleus pulposus cells with multipotent mesenchymal stromal cells from human bone marrow reveals formation of tunnelling nanotubes. Mol Med Rep. 2014;9:574–82.CrossRefPubMed
11.
Zurück zum Zitat Choi EH, Park H, Park KS, Park KS, Kim BS, Han IB, et al. Effect of nucleus pulposus cells having different phenotypes on chondrogenic differentiation of adipose-derived stromal cells in a coculture system using porous membranes. Tissue Eng Part A. 2011;17:2445–51.CrossRefPubMed Choi EH, Park H, Park KS, Park KS, Kim BS, Han IB, et al. Effect of nucleus pulposus cells having different phenotypes on chondrogenic differentiation of adipose-derived stromal cells in a coculture system using porous membranes. Tissue Eng Part A. 2011;17:2445–51.CrossRefPubMed
12.
Zurück zum Zitat Tarca AL, Romero R, Draghici S. Analysis of microarray experiments of gene expression profiling. Am J Obs Gynecol. 2006;195:373–88.CrossRef Tarca AL, Romero R, Draghici S. Analysis of microarray experiments of gene expression profiling. Am J Obs Gynecol. 2006;195:373–88.CrossRef
15.
Zurück zum Zitat Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet. 2009;10:155–9.CrossRefPubMed Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet. 2009;10:155–9.CrossRefPubMed
17.
Zurück zum Zitat Yang L, Froberg JE, Lee JT. Long noncoding RNAs: fresh perspectives into the RNA world. Trends Biochem Sci. 2014;39:35–43.CrossRefPubMed Yang L, Froberg JE, Lee JT. Long noncoding RNAs: fresh perspectives into the RNA world. Trends Biochem Sci. 2014;39:35–43.CrossRefPubMed
18.
Zurück zum Zitat Wu Q, Guo L, Jiang F, Li L, Li Z, Chen F. Analysis of the miRNA-mRNA-lncRNA networks in ER+ and ER- breast cancer cell lines. J Cell Mol Med. 2015;19:2874–87.CrossRefPubMedPubMedCentral Wu Q, Guo L, Jiang F, Li L, Li Z, Chen F. Analysis of the miRNA-mRNA-lncRNA networks in ER+ and ER- breast cancer cell lines. J Cell Mol Med. 2015;19:2874–87.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Lee CR, Sakai D, Nakai T, Toyama K, Mochida J, Alini M, et al. A phenotypic comparison of intervertebral disc and articular cartilage cells in the rat. Eur Spine J. 2007;16:2174–85.CrossRefPubMedPubMedCentral Lee CR, Sakai D, Nakai T, Toyama K, Mochida J, Alini M, et al. A phenotypic comparison of intervertebral disc and articular cartilage cells in the rat. Eur Spine J. 2007;16:2174–85.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Minogue BM, Richardson SM, Zeef LA, Freemont AJ, Hoyland JA. Transcriptional profiling of bovine intervertebral disc cells: implications for identification of normal and degenerate human intervertebral disc cell phenotypes. Arthritis Res Ther. 2010;12:R22.CrossRefPubMedPubMedCentral Minogue BM, Richardson SM, Zeef LA, Freemont AJ, Hoyland JA. Transcriptional profiling of bovine intervertebral disc cells: implications for identification of normal and degenerate human intervertebral disc cell phenotypes. Arthritis Res Ther. 2010;12:R22.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Minogue BM, Richardson SM, Zeef LAH, Freemont AJ, Hoyland JA. Characterization of the human nucleus pulposus cell phenotype and evaluation of novel marker gene expression to define adult stem cell differentiation. Arthritis Rheum. 2010;62:3695–705.CrossRefPubMed Minogue BM, Richardson SM, Zeef LAH, Freemont AJ, Hoyland JA. Characterization of the human nucleus pulposus cell phenotype and evaluation of novel marker gene expression to define adult stem cell differentiation. Arthritis Rheum. 2010;62:3695–705.CrossRefPubMed
22.
Zurück zum Zitat Rutges J, Creemers LB, Dhert W, Milz S, Sakai D, Mochida J, et al. Variations in gene and protein expression in human nucleus pulposus in comparison with annulus fibrosus and cartilage cells: potential associations with aging and degeneration. Osteoarthr Cartil. 2010;18:416–23.CrossRefPubMed Rutges J, Creemers LB, Dhert W, Milz S, Sakai D, Mochida J, et al. Variations in gene and protein expression in human nucleus pulposus in comparison with annulus fibrosus and cartilage cells: potential associations with aging and degeneration. Osteoarthr Cartil. 2010;18:416–23.CrossRefPubMed
23.
Zurück zum Zitat Xi Y, Jiang T, Wang W, Yu J, Wang Y, Wu X, et al. Long non-coding HCG18 promotes intervertebral disc degeneration by sponging miR-146a-5p and regulating TRAF6 expression. Sci Rep. 2017;7:13234.CrossRefPubMedPubMedCentral Xi Y, Jiang T, Wang W, Yu J, Wang Y, Wu X, et al. Long non-coding HCG18 promotes intervertebral disc degeneration by sponging miR-146a-5p and regulating TRAF6 expression. Sci Rep. 2017;7:13234.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Li Z, Yu X, Shen J, Chan MT, Wu WK. MicroRNA in intervertebral disc degeneration. Cell Prolif. 2015;48:278–83.CrossRefPubMed Li Z, Yu X, Shen J, Chan MT, Wu WK. MicroRNA in intervertebral disc degeneration. Cell Prolif. 2015;48:278–83.CrossRefPubMed
26.
Zurück zum Zitat Yan N, Yu S, Zhang H, Hou T. Lumbar disc degeneration is facilitated by MiR-100-mediated FGFR3 suppression. Cell Physiol Biochem. 2015;36:2229–36.CrossRefPubMed Yan N, Yu S, Zhang H, Hou T. Lumbar disc degeneration is facilitated by MiR-100-mediated FGFR3 suppression. Cell Physiol Biochem. 2015;36:2229–36.CrossRefPubMed
27.
Zurück zum Zitat Li H, Cui Q, Dong ZY, Zhang JH, Li H, Zhao L. Downregulation of miR-27b is involved in loss of type ii collagen by directly targeting matrix metalloproteinase 13 (MMP13) in human intervertebral disc degeneration. Spine (Phila Pa 1976). 2016;41:E116–23.CrossRef Li H, Cui Q, Dong ZY, Zhang JH, Li H, Zhao L. Downregulation of miR-27b is involved in loss of type ii collagen by directly targeting matrix metalloproteinase 13 (MMP13) in human intervertebral disc degeneration. Spine (Phila Pa 1976). 2016;41:E116–23.CrossRef
28.
Zurück zum Zitat Jing W, Jiang W. MicroRNA-93 regulates collagen loss by targeting MMP3 in human nucleus pulposus cells. Cell Prolif. 2015;48:284–92.CrossRefPubMed Jing W, Jiang W. MicroRNA-93 regulates collagen loss by targeting MMP3 in human nucleus pulposus cells. Cell Prolif. 2015;48:284–92.CrossRefPubMed
29.
Zurück zum Zitat Pfirrmann CW, Metzdorf A, Zanetti M, Hodler J, Boos N. Magnetic resonance classification of lumbar intervertebral disc degeneration. Spine (Phila Pa 1976). 2001;26:1873–8.CrossRef Pfirrmann CW, Metzdorf A, Zanetti M, Hodler J, Boos N. Magnetic resonance classification of lumbar intervertebral disc degeneration. Spine (Phila Pa 1976). 2001;26:1873–8.CrossRef
30.
Zurück zum Zitat Ruan D, Zhang Y, Wang D, Zhang C, Wu J, Wang C, et al. Differentiation of human Wharton’s jelly cells toward nucleus pulposus-like cells after coculture with nucleus pulposus cells in vitro. Tissue Eng Part A. 2012;18:167–75.CrossRefPubMed Ruan D, Zhang Y, Wang D, Zhang C, Wu J, Wang C, et al. Differentiation of human Wharton’s jelly cells toward nucleus pulposus-like cells after coculture with nucleus pulposus cells in vitro. Tissue Eng Part A. 2012;18:167–75.CrossRefPubMed
31.
Zurück zum Zitat Russo V, Yu C, Belliveau P, Hamilton A, Flynn LE. Comparison of human adipose-derived stem cells isolated from subcutaneous, omental, and intrathoracic adipose tissue depots for regenerative applications. Stem Cells Transl Med. 2014;3:206–17.CrossRefPubMed Russo V, Yu C, Belliveau P, Hamilton A, Flynn LE. Comparison of human adipose-derived stem cells isolated from subcutaneous, omental, and intrathoracic adipose tissue depots for regenerative applications. Stem Cells Transl Med. 2014;3:206–17.CrossRefPubMed
32.
Zurück zum Zitat Schaffler A, Buchler C. Concise review: adipose tissue-derived stromal cells–basic and clinical implications for novel cell-based therapies. Stem Cells. 2007;25:818–27.CrossRefPubMed Schaffler A, Buchler C. Concise review: adipose tissue-derived stromal cells–basic and clinical implications for novel cell-based therapies. Stem Cells. 2007;25:818–27.CrossRefPubMed
33.
Zurück zum Zitat Chen J, Xu J, Li Y, Zhang J, Chen H, Lu J, et al. Competing endogenous RNA network analysis identifies critical genes among the different breast cancer subtypes. Oncotarget. 2017;8:10171–84.PubMed Chen J, Xu J, Li Y, Zhang J, Chen H, Lu J, et al. Competing endogenous RNA network analysis identifies critical genes among the different breast cancer subtypes. Oncotarget. 2017;8:10171–84.PubMed
34.
Zurück zum Zitat Jonas S, Izaurralde E. Towards a molecular understanding of microRNA-mediated gene silencing. Nat Rev Genet. 2015;16:421–33.CrossRefPubMed Jonas S, Izaurralde E. Towards a molecular understanding of microRNA-mediated gene silencing. Nat Rev Genet. 2015;16:421–33.CrossRefPubMed
36.
Zurück zum Zitat Li CY, Liang GY, Yao WZ, Sui J, Shen X, Zhang YQ, et al. Integrated analysis of long non-coding RNA competing interactions reveals the potential role in progression of human gastric cancer. Int J Oncol. 2016;48:1965–76.CrossRefPubMed Li CY, Liang GY, Yao WZ, Sui J, Shen X, Zhang YQ, et al. Integrated analysis of long non-coding RNA competing interactions reveals the potential role in progression of human gastric cancer. Int J Oncol. 2016;48:1965–76.CrossRefPubMed
37.
Zurück zum Zitat Le Visage C, Kim SW, Tateno K, Sieber AN, Kostuik JP, Leong KW. Interaction of human mesenchymal stem cells with disc cells: changes in extracellular matrix biosynthesis. Spine (Phila Pa 1976). 2006;31:2036–42.CrossRef Le Visage C, Kim SW, Tateno K, Sieber AN, Kostuik JP, Leong KW. Interaction of human mesenchymal stem cells with disc cells: changes in extracellular matrix biosynthesis. Spine (Phila Pa 1976). 2006;31:2036–42.CrossRef
38.
Zurück zum Zitat Wei AQ, Chung SA, Tao H, Brisby H, Lin Z, Shen BJ, et al. Differentiation of rodent bone marrow mesenchymal stem cells into intervertebral disc-like cells following coculture with rat disc tissue. Tissue Eng Part A. 2009;15:2581–93.CrossRefPubMed Wei AQ, Chung SA, Tao H, Brisby H, Lin Z, Shen BJ, et al. Differentiation of rodent bone marrow mesenchymal stem cells into intervertebral disc-like cells following coculture with rat disc tissue. Tissue Eng Part A. 2009;15:2581–93.CrossRefPubMed
39.
Zurück zum Zitat Richardson SM, Walker RV, Parker S, Rhodes NP, Hunt JA, Freemont AJ, et al. Intervertebral disc cell-mediated mesenchymal stem cell differentiation. Stem Cells. 2006;24:707–16.CrossRefPubMed Richardson SM, Walker RV, Parker S, Rhodes NP, Hunt JA, Freemont AJ, et al. Intervertebral disc cell-mediated mesenchymal stem cell differentiation. Stem Cells. 2006;24:707–16.CrossRefPubMed
40.
Zurück zum Zitat Niu CC, Yuan LJ, Lin SS, Chen LH, Chen WJ. Mesenchymal stem cell and nucleus pulposus cell coculture modulates cell profile. Clin Orthop Relat Res. 2009;467:3263–72.CrossRefPubMed Niu CC, Yuan LJ, Lin SS, Chen LH, Chen WJ. Mesenchymal stem cell and nucleus pulposus cell coculture modulates cell profile. Clin Orthop Relat Res. 2009;467:3263–72.CrossRefPubMed
42.
Zurück zum Zitat Marfia G, Navone SE, Di Vito C, Tabano S, Giammattei L, Di Cristofori A, et al. Gene expression profile analysis of human mesenchymal stem cells from herniated and degenerated intervertebral discs reveals different expression of osteopontin. Stem Cells Dev. 2015;24:320–8.CrossRefPubMed Marfia G, Navone SE, Di Vito C, Tabano S, Giammattei L, Di Cristofori A, et al. Gene expression profile analysis of human mesenchymal stem cells from herniated and degenerated intervertebral discs reveals different expression of osteopontin. Stem Cells Dev. 2015;24:320–8.CrossRefPubMed
43.
Zurück zum Zitat Power KA, Grad S, Rutges JPHJ, Creemers LB, van Rijen MHP, O’Gaora P, et al. Identification of cell surface-specific markers to target human nucleus pulposus cells: expression of carbonic anhydrase XII varies with age and degeneration. Arthritis Rheum. 2011;63:3876–86.CrossRefPubMed Power KA, Grad S, Rutges JPHJ, Creemers LB, van Rijen MHP, O’Gaora P, et al. Identification of cell surface-specific markers to target human nucleus pulposus cells: expression of carbonic anhydrase XII varies with age and degeneration. Arthritis Rheum. 2011;63:3876–86.CrossRefPubMed
44.
Zurück zum Zitat Rodrigues-Pinto R, Richardson SM, Hoyland JA. Identification of novel nucleus pulposus markers: interspecies variations and implications for cell-based therapies for intervertebral disc degeneration. Bone Joint Res. 2013;2:169–78.CrossRefPubMedPubMedCentral Rodrigues-Pinto R, Richardson SM, Hoyland JA. Identification of novel nucleus pulposus markers: interspecies variations and implications for cell-based therapies for intervertebral disc degeneration. Bone Joint Res. 2013;2:169–78.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Risbud MV, Schoepflin ZR, Mwale F, Kandel RA, Grad S, Iatridis JC, et al. Defining the phenotype of young healthy nucleus pulposus cells: recommendations of the Spine Research Interest Group at the 2014 annual ORS meeting. J Orthop Res. 2015;33:283–93.CrossRefPubMedPubMedCentral Risbud MV, Schoepflin ZR, Mwale F, Kandel RA, Grad S, Iatridis JC, et al. Defining the phenotype of young healthy nucleus pulposus cells: recommendations of the Spine Research Interest Group at the 2014 annual ORS meeting. J Orthop Res. 2015;33:283–93.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Fujita N, Miyamoto T, Imai J, Hosogane N, Suzuki T, Yagi M, et al. CD24 is expressed specifically in the nucleus pulposus of intervertebral discs. Biochem Biophys Res Commun. 2005;338:1890–6.CrossRefPubMed Fujita N, Miyamoto T, Imai J, Hosogane N, Suzuki T, Yagi M, et al. CD24 is expressed specifically in the nucleus pulposus of intervertebral discs. Biochem Biophys Res Commun. 2005;338:1890–6.CrossRefPubMed
47.
Zurück zum Zitat Rodrigues-Pinto R, Berry A, Piper-Hanley K, Hanley N, Richardson SM, Hoyland JA. Spatiotemporal analysis of putative notochordal cell markers reveals CD24 and keratins 8, 18, and 19 as notochord-specific markers during early human intervertebral disc development. J Orthop Res. 2016;34:1327–40.CrossRefPubMedPubMedCentral Rodrigues-Pinto R, Berry A, Piper-Hanley K, Hanley N, Richardson SM, Hoyland JA. Spatiotemporal analysis of putative notochordal cell markers reveals CD24 and keratins 8, 18, and 19 as notochord-specific markers during early human intervertebral disc development. J Orthop Res. 2016;34:1327–40.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Tang R, Jing L, Willard VP, Wu C-L, Guilak F, Chen J, et al. Differentiation of human induced pluripotent stem cells into nucleus pulposus-like cells. Stem Cell Res Ther. 2018;9:61.CrossRefPubMedPubMedCentral Tang R, Jing L, Willard VP, Wu C-L, Guilak F, Chen J, et al. Differentiation of human induced pluripotent stem cells into nucleus pulposus-like cells. Stem Cell Res Ther. 2018;9:61.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Sakai D, Nakamura Y, Nakai T, Mishima T, Kato S, Grad S, et al. Exhaustion of nucleus pulposus progenitor cells with ageing and degeneration of the intervertebral disc. Nat Commun. 2012;3:1264.CrossRefPubMedPubMedCentral Sakai D, Nakamura Y, Nakai T, Mishima T, Kato S, Grad S, et al. Exhaustion of nucleus pulposus progenitor cells with ageing and degeneration of the intervertebral disc. Nat Commun. 2012;3:1264.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Gumbiner BM. Cell adhesion: the molecular basis of tissue architecture and morphogenesis. Cell. 1996;84:345–57.CrossRefPubMed Gumbiner BM. Cell adhesion: the molecular basis of tissue architecture and morphogenesis. Cell. 1996;84:345–57.CrossRefPubMed
51.
Zurück zum Zitat Roeder RG. The role of general initiation factors in transcription by RNA polymerase II. Trends Biochem Sci. 1996;21:327–35.CrossRefPubMed Roeder RG. The role of general initiation factors in transcription by RNA polymerase II. Trends Biochem Sci. 1996;21:327–35.CrossRefPubMed
52.
Zurück zum Zitat Maidhof R, Alipui DO, Rafiuddin A, Levine M, Grande DA, Chahine NO. Emerging trends in biological therapy for intervertebral disc degeneration. Discov Med. 2012;14:401–11.PubMed Maidhof R, Alipui DO, Rafiuddin A, Levine M, Grande DA, Chahine NO. Emerging trends in biological therapy for intervertebral disc degeneration. Discov Med. 2012;14:401–11.PubMed
53.
Zurück zum Zitat Wuertz K, Vo N, Kletsas D, Boos N. Inflammatory and catabolic signalling in intervertebral discs: the roles of NF-kappaB and MAP kinases. Eur Cell Mater. 2012;23:103–20.PubMed Wuertz K, Vo N, Kletsas D, Boos N. Inflammatory and catabolic signalling in intervertebral discs: the roles of NF-kappaB and MAP kinases. Eur Cell Mater. 2012;23:103–20.PubMed
54.
Zurück zum Zitat Tian Y, Yuan W, Fujita N, Wang J, Wang H, Shapiro IM, et al. Inflammatory cytokines associated with degenerative disc disease control aggrecanase-1 (ADAMTS-4) expression in nucleus pulposus cells through MAPK and NF-kappaB. Am J Pathol. 2013;182:2310–21.CrossRefPubMedPubMedCentral Tian Y, Yuan W, Fujita N, Wang J, Wang H, Shapiro IM, et al. Inflammatory cytokines associated with degenerative disc disease control aggrecanase-1 (ADAMTS-4) expression in nucleus pulposus cells through MAPK and NF-kappaB. Am J Pathol. 2013;182:2310–21.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Liu C, Yang H, Gao F, Li X, An Y, Wang J, et al. Resistin promotes intervertebral disc degeneration by upregulation of ADAMTS-5 through p38 MAPK signaling pathway. Spine (Phila Pa 1976). 2016;41:1414–20.CrossRef Liu C, Yang H, Gao F, Li X, An Y, Wang J, et al. Resistin promotes intervertebral disc degeneration by upregulation of ADAMTS-5 through p38 MAPK signaling pathway. Spine (Phila Pa 1976). 2016;41:1414–20.CrossRef
56.
Zurück zum Zitat Niu CC, Lin SS, Yuan LJ, Chen LH, Wang IC, Tsai TT, et al. Hyperbaric oxygen treatment suppresses MAPK signaling and mitochondrial apoptotic pathway in degenerated human intervertebral disc cells. J Orthop Res. 2013;31:204–9.CrossRefPubMed Niu CC, Lin SS, Yuan LJ, Chen LH, Wang IC, Tsai TT, et al. Hyperbaric oxygen treatment suppresses MAPK signaling and mitochondrial apoptotic pathway in degenerated human intervertebral disc cells. J Orthop Res. 2013;31:204–9.CrossRefPubMed
57.
Zurück zum Zitat Kletsas D. Senescent cells in the intervertebral disc: numbers and mechanisms. Spine J. 2009;9:677–8.CrossRefPubMed Kletsas D. Senescent cells in the intervertebral disc: numbers and mechanisms. Spine J. 2009;9:677–8.CrossRefPubMed
58.
Zurück zum Zitat Gruber HE, Hoelscher GL, Ingram JA, Zinchenko N, Hanley EN Jr. Senescent vs. non-senescent cells in the human annulus in vivo: cell harvest with laser capture microdissection and gene expression studies with microarray analysis. BMC Biotechnol. 2010;10:5.CrossRefPubMedPubMedCentral Gruber HE, Hoelscher GL, Ingram JA, Zinchenko N, Hanley EN Jr. Senescent vs. non-senescent cells in the human annulus in vivo: cell harvest with laser capture microdissection and gene expression studies with microarray analysis. BMC Biotechnol. 2010;10:5.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Adams MA, Stefanakis M, Dolan P. Healing of a painful intervertebral disc should not be confused with reversing disc degeneration: implications for physical therapies for discogenic back pain. Clin Biomech (Bristol, Avon). 2010;25:961–71.CrossRef Adams MA, Stefanakis M, Dolan P. Healing of a painful intervertebral disc should not be confused with reversing disc degeneration: implications for physical therapies for discogenic back pain. Clin Biomech (Bristol, Avon). 2010;25:961–71.CrossRef
60.
Zurück zum Zitat Hoyland JA, Le Maitre C, Freemont AJ. Investigation of the role of IL-1 and TNF in matrix degradation in the intervertebral disc. Rheumatol. 2008;47:809–14.CrossRef Hoyland JA, Le Maitre C, Freemont AJ. Investigation of the role of IL-1 and TNF in matrix degradation in the intervertebral disc. Rheumatol. 2008;47:809–14.CrossRef
61.
Zurück zum Zitat Le Maitre CL, Hoyland JA, Freemont AJ. Catabolic cytokine expression in degenerate and herniated human intervertebral discs: IL-1beta and TNFalpha expression profile. Arthritis Res Ther. 2007;9:R77.CrossRefPubMedPubMedCentral Le Maitre CL, Hoyland JA, Freemont AJ. Catabolic cytokine expression in degenerate and herniated human intervertebral discs: IL-1beta and TNFalpha expression profile. Arthritis Res Ther. 2007;9:R77.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Ouyang ZH, Wang WJ, Yan YG, Wang B, Lv GH. The PI3K/Akt pathway: a critical player in intervertebral disc degeneration. Oncotarget. 2017;8:57870–81.PubMedPubMedCentral Ouyang ZH, Wang WJ, Yan YG, Wang B, Lv GH. The PI3K/Akt pathway: a critical player in intervertebral disc degeneration. Oncotarget. 2017;8:57870–81.PubMedPubMedCentral
63.
Zurück zum Zitat Liu Z, Zhou K, Fu W, Zhang H. Insulin-like growth factor 1 activates PI3k/Akt signaling to antagonize lumbar disc degeneration. Cell Physiol Biochem. 2015;37:225–32.CrossRefPubMed Liu Z, Zhou K, Fu W, Zhang H. Insulin-like growth factor 1 activates PI3k/Akt signaling to antagonize lumbar disc degeneration. Cell Physiol Biochem. 2015;37:225–32.CrossRefPubMed
64.
Zurück zum Zitat Zhang M, Zhou Q, Liang QQ, Li CG, Holz JD, Tang D, et al. IGF-1 regulation of type II collagen and MMP-13 expression in rat endplate chondrocytes via distinct signaling pathways. Osteoarthr Cartil. 2009;17:100–6.CrossRefPubMed Zhang M, Zhou Q, Liang QQ, Li CG, Holz JD, Tang D, et al. IGF-1 regulation of type II collagen and MMP-13 expression in rat endplate chondrocytes via distinct signaling pathways. Osteoarthr Cartil. 2009;17:100–6.CrossRefPubMed
65.
Zurück zum Zitat Pratsinis H, Constantinou V, Pavlakis K, Sapkas G, Kletsas D. Exogenous and autocrine growth factors stimulate human intervertebral disc cell proliferation via the ERK and Akt pathways. J Orthop Res. 2012;30:958–64.CrossRefPubMed Pratsinis H, Constantinou V, Pavlakis K, Sapkas G, Kletsas D. Exogenous and autocrine growth factors stimulate human intervertebral disc cell proliferation via the ERK and Akt pathways. J Orthop Res. 2012;30:958–64.CrossRefPubMed
66.
Zurück zum Zitat Wang D, Hu Z, Hao J, He B, Gan Q, Zhong X, et al. SIRT1 inhibits apoptosis of degenerative human disc nucleus pulposus cells through activation of Akt pathway. Age. 2013;35:1741–53.CrossRefPubMed Wang D, Hu Z, Hao J, He B, Gan Q, Zhong X, et al. SIRT1 inhibits apoptosis of degenerative human disc nucleus pulposus cells through activation of Akt pathway. Age. 2013;35:1741–53.CrossRefPubMed
67.
Zurück zum Zitat Krupkova O, Handa J, Hlavna M, Klasen J, Ospelt C, Ferguson SJ, et al. The natural polyphenol epigallocatechin Gallate protects intervertebral disc cells from oxidative stress. Oxidative Med Cell Longev. 2016;2016:7031397. Krupkova O, Handa J, Hlavna M, Klasen J, Ospelt C, Ferguson SJ, et al. The natural polyphenol epigallocatechin Gallate protects intervertebral disc cells from oxidative stress. Oxidative Med Cell Longev. 2016;2016:​7031397.
68.
Zurück zum Zitat Doble BW, Woodgett JR. GSK-3: tricks of the trade for a multi-tasking kinase. J Cell Sci. 2003;116:1175–86. Doble BW, Woodgett JR. GSK-3: tricks of the trade for a multi-tasking kinase. J Cell Sci. 2003;116:1175–86.
69.
Zurück zum Zitat Patel S, Doble B, Woodgett JR. Glycogen synthase kinase-3 in insulin and Wnt signalling: a double-edged sword? Biochem Soc Trans. 2004;32:803–8.CrossRefPubMedPubMedCentral Patel S, Doble B, Woodgett JR. Glycogen synthase kinase-3 in insulin and Wnt signalling: a double-edged sword? Biochem Soc Trans. 2004;32:803–8.CrossRefPubMedPubMedCentral
70.
Zurück zum Zitat Kita K, Kimura T, Nakamura N, Yoshikawa H, Nakano T. PI3K/Akt signaling as a key regulatory pathway for chondrocyte terminal differentiation. Genes Cells. 2008;13:839–50.CrossRefPubMed Kita K, Kimura T, Nakamura N, Yoshikawa H, Nakano T. PI3K/Akt signaling as a key regulatory pathway for chondrocyte terminal differentiation. Genes Cells. 2008;13:839–50.CrossRefPubMed
71.
Zurück zum Zitat Li TF, Zuscik MJ, Ionescu AM, Zhang X, Rosier RN, Schwarz EM, et al. PGE2 inhibits chondrocyte differentiation through PKA and PKC signaling. Exp Cell Res. 2004;300:159–69.CrossRefPubMed Li TF, Zuscik MJ, Ionescu AM, Zhang X, Rosier RN, Schwarz EM, et al. PGE2 inhibits chondrocyte differentiation through PKA and PKC signaling. Exp Cell Res. 2004;300:159–69.CrossRefPubMed
72.
Zurück zum Zitat Day TF, Guo X, Garrett-Beal L, Yang Y. Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev Cell. 2005;8:739–50.CrossRefPubMed Day TF, Guo X, Garrett-Beal L, Yang Y. Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev Cell. 2005;8:739–50.CrossRefPubMed
73.
Zurück zum Zitat Miclea RL, Siebelt M, Finos L, Goeman JJ, Lowik CW, Oostdijk W, et al. Inhibition of Gsk3beta in cartilage induces osteoarthritic features through activation of the canonical Wnt signaling pathway. Osteoarthr Cartil. 2011;19:1363–72.CrossRefPubMed Miclea RL, Siebelt M, Finos L, Goeman JJ, Lowik CW, Oostdijk W, et al. Inhibition of Gsk3beta in cartilage induces osteoarthritic features through activation of the canonical Wnt signaling pathway. Osteoarthr Cartil. 2011;19:1363–72.CrossRefPubMed
74.
Zurück zum Zitat Itoh S, Saito T, Hirata M, Ushita M, Ikeda T, Woodgett JR, et al. GSK-3alpha and GSK-3beta proteins are involved in early stages of chondrocyte differentiation with functional redundancy through RelA protein phosphorylation. J Biol Chem. 2012;287:29227–36.CrossRefPubMedPubMedCentral Itoh S, Saito T, Hirata M, Ushita M, Ikeda T, Woodgett JR, et al. GSK-3alpha and GSK-3beta proteins are involved in early stages of chondrocyte differentiation with functional redundancy through RelA protein phosphorylation. J Biol Chem. 2012;287:29227–36.CrossRefPubMedPubMedCentral
75.
Zurück zum Zitat Brockdorff N, Ashworth A, Kay GF, McCabe VM, Norris DP, Cooper PJ, et al. The product of the mouse Xist gene is a 15 kb inactive X-specific transcript containing no conserved ORF and located in the nucleus. Cell. 1992;71:515–26.CrossRefPubMed Brockdorff N, Ashworth A, Kay GF, McCabe VM, Norris DP, Cooper PJ, et al. The product of the mouse Xist gene is a 15 kb inactive X-specific transcript containing no conserved ORF and located in the nucleus. Cell. 1992;71:515–26.CrossRefPubMed
76.
Zurück zum Zitat Brown CJ, Hendrich BD, Rupert JL, Lafreniere RG, Xing Y, Lawrence J, et al. The human XIST gene: analysis of a 17 kb inactive X-specific RNA that contains conserved repeats and is highly localized within the nucleus. Cell. 1992;71:527–42.CrossRefPubMed Brown CJ, Hendrich BD, Rupert JL, Lafreniere RG, Xing Y, Lawrence J, et al. The human XIST gene: analysis of a 17 kb inactive X-specific RNA that contains conserved repeats and is highly localized within the nucleus. Cell. 1992;71:527–42.CrossRefPubMed
77.
Zurück zum Zitat Clemson CM, McNeil JA, Willard HF, Lawrence JB. XIST RNA paints the inactive X chromosome at interphase: evidence for a novel RNA involved in nuclear/chromosome structure. J Cell Biol. 1996;132:259–75.CrossRefPubMed Clemson CM, McNeil JA, Willard HF, Lawrence JB. XIST RNA paints the inactive X chromosome at interphase: evidence for a novel RNA involved in nuclear/chromosome structure. J Cell Biol. 1996;132:259–75.CrossRefPubMed
78.
Zurück zum Zitat Penny GD, Kay GF, Sheardown SA, Rastan S, Brockdorff N. Requirement for Xist in X chromosome inactivation. Nature. 1996;379:131–7.CrossRefPubMed Penny GD, Kay GF, Sheardown SA, Rastan S, Brockdorff N. Requirement for Xist in X chromosome inactivation. Nature. 1996;379:131–7.CrossRefPubMed
79.
Zurück zum Zitat Lee JT, Bartolomei MS. X-inactivation, imprinting, and long noncoding RNAs in health and disease. Cell. 2013;152:1308–23.CrossRefPubMed Lee JT, Bartolomei MS. X-inactivation, imprinting, and long noncoding RNAs in health and disease. Cell. 2013;152:1308–23.CrossRefPubMed
80.
Zurück zum Zitat Minkovsky A, Patel S, Plath K. Concise review: pluripotency and the transcriptional inactivation of the female mammalian X chromosome. Stem Cells. 2012;30:48–54.CrossRefPubMedPubMedCentral Minkovsky A, Patel S, Plath K. Concise review: pluripotency and the transcriptional inactivation of the female mammalian X chromosome. Stem Cells. 2012;30:48–54.CrossRefPubMedPubMedCentral
81.
Zurück zum Zitat Ji ML, Lu J, Shi PL, Zhang XJ, Wang SZ, Chang Q, et al. Dysregulated miR-98 contributes to extracellular matrix degradation by targeting IL-6/STAT3 signaling pathway in human intervertebral disc degeneration. J Bone Min Res. 2016;31:900–9.CrossRef Ji ML, Lu J, Shi PL, Zhang XJ, Wang SZ, Chang Q, et al. Dysregulated miR-98 contributes to extracellular matrix degradation by targeting IL-6/STAT3 signaling pathway in human intervertebral disc degeneration. J Bone Min Res. 2016;31:900–9.CrossRef
Metadaten
Titel
Aberrantly expressed messenger RNAs and long noncoding RNAs in degenerative nucleus pulposus cells co-cultured with adipose-derived mesenchymal stem cells
verfasst von
Zhihua Han
Jiandong Wang
Liang Gao
Qiugen Wang
Jianhong Wu
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Arthritis Research & Therapy / Ausgabe 1/2018
Elektronische ISSN: 1478-6362
DOI
https://doi.org/10.1186/s13075-018-1677-x

Weitere Artikel der Ausgabe 1/2018

Arthritis Research & Therapy 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Strenge Blutdruckeinstellung lohnt auch im Alter noch

30.04.2024 Arterielle Hypertonie Nachrichten

Ältere Frauen, die von chronischen Erkrankungen weitgehend verschont sind, haben offenbar die besten Chancen, ihren 90. Geburtstag zu erleben, wenn ihr systolischer Blutdruck < 130 mmHg liegt. Das scheint selbst für 80-Jährige noch zu gelten.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.