Skip to main content
Erschienen in: BMC Musculoskeletal Disorders 1/2021

Open Access 01.12.2021 | Research article

Major ceRNA regulation and key metabolic signature analysis of intervertebral disc degeneration

verfasst von: Shuai Cao, Jie Li, Kai Yang, Haopeng Li

Erschienen in: BMC Musculoskeletal Disorders | Ausgabe 1/2021

Abstract

Background and objective

Intervertebral disc degeneration (IDD) is a complex multifactorial and irreversible pathological process. In IDD, multiple competing endogenous RNAs (ceRNA, including mRNA, lncRNA, and pseudogenes) can compete to bind with miRNAs. However, the potential metabolic signatures in nucleus pulposus (NP) cells remain poorly understood. This study investigated key metabolic genes and the ceRNA regulatory mechanisms in the pathogenesis of IDD based on microarray datasets.

Methods

We retrieved and downloaded four independent IDD microarray datasets from the Gene Expression Omnibus. Combining the predicted interactions from online databases (miRcode, miRDB, miRTarBase, and TargetScan), differentially expressed lncRNAs (DElncRNAs), miRNAs (DEmiRNAs), and mRNAs (DEmRNAs) were identified. A ceRNA network was constructed and annotated using GO and KEGG pathway enrichment analyses. Moreover, we searched the online metabolic gene set and used support vector machine (SVM) to find the critical metabolic DEmRNA(s) and other DERNAs. Differential gene expression was validated with a merged dataset.

Results

A total of 45 DEmRNAs, 36 DElncRNAs, and only one DEmiRNA (miR-338-3p) were identified in the IDD microarray datasets. GO and KEGG pathway enrichment analyses revealed that the DEmRNAs were predominantly enriched in the PI3K-Akt signaling pathway, MAPK signaling pathway, IL-17 signaling pathway, apoptosis, and cellular response to oxidative stress. Based on SVM screening, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFK/FBPase) 2 is the critical metabolic gene with lower expression in IDD, and AC063977.6 is the key lncRNA with lower expression in IDD. The ceRNA hypothesis suggests that AC063977.6, miR-338-3p (high expression), and PFKFB2 are dysregulated as an axis in IDD.

Conclusions

The results suggest that lncRNA AC063977.6 correlate with PFKFB2, the vital metabolic signature gene, via targeting miR-338-3p during IDD pathogenesis. The current study may shed light on unraveling the pathogenesis of IDD.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12891-021-04109-8.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
IDD
Intervertebral disc degeneration
miRNAs
MicroRNAs
lncRNAs
Long noncoding RNAs
ceRNA
Competing endogenous RNA
NP
Nucleus pulposus
GEO
Gene Expression Omnibus
DE
Differential expression
DElncRNAs
Differentially expressed lncRNAs
DEmiRNAs
Differentially expressed miRNAs
DEmRNAs
Differentially expressed mRNAs
SVM
Support vector machine
PFKFB
6-phosphofructo-2-kinase/fructose-2,6-bisphosphatas
AUC
The area under curve
LBP
Low-back pain
ECM
Extracellular matrix
mRNA
Messenger RNA
DEGs
Differentially expressed genes
GO
Gene Ontology
KEGG
Kyoto Encyclopedia of Genes and Genomes
ROC
Receiver operating characteristic

Background

Intervertebral disc degeneration (IDD), a complex multifactorial and age-dependent condition, is a critical contributor to low-back pain (LBP) [1]. The increasing incidence of IDD not only affects the life quality of patients because of chronic pain and disability, but also causes a severe financial burden to the health care system [2]. Moreover, current prevention, diagnosis, and treatment modalities for IDD are hampered due to its ambiguous pathogenic mechanisms. Previous studies have shown that IDD is characterized by the abnormal proliferation of nucleus pulposus (NP) cells, degradation of proteoglycans and collagen in the extracellular matrix (ECM), imbalance of homeostasis, and accelerated transition of NP cells from a healthy state to a catabolic and degenerative state [3]. It is known that the cellular function of all life forms relies closely on metabolism [4, 5]. Metabolic genes are believed to play essential roles in various cellular functions and many diseases such as hepatic fibrosis, central nervous system diseases, and hematologic malignancies [68]. However, investigations concerning the role of metabolic networks in IDD remain sparse. Hence, in-depth comprehension of metabolic gene expression and the regulatory mechanisms of noncoding RNA may provide novel insights into the onset, development, and diagnosis of IDD.
Long noncoding RNAs (lncRNAs), a group of RNA molecules consisting of more than 200 nucleotides with no or feeble protein-coding ability [3], have been of great interest because of their ability to regulate gene expression in various pathological and biological processes, such as cell proliferation and apoptosis [9, 10]. MicroRNAs (miRNAs), consisting of approximately 18–22 nucleotides without coding function, mediate post-transcriptional regulation of target messenger RNA (mRNA) [11]. Ample studies indicate that lncRNAs can act as natural microRNA (miRNA) sponges to bind miRNAs, acting as competing endogenous RNAs (ceRNAs) [1214]. The ceRNA crosstalk plays a critical role in modulating gene expression [15]. Accumulating evidence has demonstrated the aberrant expression of several mRNAs, miRNAs, and lncRNAs in NP cells in IDD and explained the relationship between them with respect to autophagy, apoptosis, and cell cycle [11, 16, 17]. Nevertheless, metabolic gene-associated ceRNA regulated IDD progression remains largely unstudied. Hence, further exploration of metabolism-related ceRNA crosstalk in NP cells will benefit our understanding of the metabolic gene regulatory network, which is of considerable significance for understanding IDD pathogenic mechanisms. Besides, it is vital to identify the key metabolic gene(s) and relevant key regulated noncoding RNAs in the IDD. In addition, we previously screened core RNAs of IDD based not only on differential expression (DE) but also using the machine learning technology, namely support vector machine (SVM) [18].
Currently, the definitive diagnosis of IDD is sometimes difficult because the intervertebral disc has a particular anatomical structure. In addition, LBP usually has atypical clinical features [19], and the definitive diagnosis is not often achieved using clinical imaging methods [20]. It is even more difficult to correctly diagnose dorsal disc migrations [21], and misdiagnosis often occurs [22]. A transcriptomic signature is beneficial for clear and early diagnosis and timely treatment of patients with IDD [23], especially those that are at a high risk of IDD. Currently, the metabolic gene signature is a collection of metabolism-related genes that can characterize the outcome events; it accounts for the top differentially regulated genes in many disorders but is relatively poorly explored in IDD [24, 25]. In the current study, we aimed to explore the key metabolic gene(s) signature and its key ceRNA regulatory systems to provide insight into IDD pathogenesis. Combining database prediction and gene expression validation, we explored the metabolic abnormalities of NP cells in IDD and revealed their underlying metabolic, molecular signatures and regulation profiles based on microarray datasets.

Methods

Data acquisition and study design (Fig. 1)

After searching the National Center of Biotechnology Information (NCBI) Gene Expression Omnibus (GEO) database (https://​www.​ncbi.​nlm.​nih.​gov/​geo), we retrieved and downloaded all four IDD microarray datasets (lncRNA microarray set, GSE56081; miRNA microarray sets GSE116726 and GSE19943; and mRNA microarray sets, GSE56081 and GSE70362). According to the Thompson grading system, grade I to grade III NP samples were classified as control samples, and grade IV and grade V NP samples were classified as IDD samples [26]. GSE56081 consists of five IDD and five control samples, whereas GSE116726 and GSE19943 include three IDD and three control samples, respectively. The GSE70362 consists of 14 control samples and 10 IDD samples (Table 1). In order to obtain standardized data, we performed batch normalization on GSE116726 and GSE19943 using the R package ‘sva’. For mRNA analysis, the GSE56081 microarray set was used for initial exploration, and the GSE70362 microarray set was normalized, merged and used for further validation (batch normalization and merging with GSE56081 to obtain large sample size). Because the data were publicly obtained from the GEO database, we strictly followed the publication guidelines approved by GEO (details with platform information, sample size, and access are shown in Supplementary Table 1. The characteristics of IDD patients in related GEO datasets are shown in Supplementary Table 2). And, ethics committee approval was not required to conduct this study.
Table 1
Details and sample size with studied GEO datasets
Datasets
Platform information
Control(N)
IDD(N)
Application
GSE56081
GPL15314 Arraystar Human LncRNA microarray V2.0
5
5
identification of DElncRNA
GSE116726
GPL20712 Agilent-070156 Human miRNA [miRNA version]
3
3
identification of DEmiRNA
GSE19943
GPL9946 Exiqon human miRCURY LNA™ miRNA Array V11.0
3
3
identification of DEmiRNA
GSE56081
GPL15314 Arraystar Human LncRNA microarray V2.0
5
5
identification of metabolic DEGs
GSE70362
GPL17810[HG-U133_Plus_2] Affymetrix Human Genome U133
14
10
validation of metabolic DEGs
Abbreviations: GEO Gene Expression Omnibus, IDD Intervertebral disc degeneration, DElncRNAs differentially expressed lncRNAs, DEmiRNAs differentially expressed miRNAs, DEGs differentially expressed genes

DE lncRNAs, miRNAs, mRNA screening

The ceRNA network was constructed based on DE analysis for differentially expressed lncRNAs (DElncRNAs), differentially expressed miRNAs (DEmiRNAs) and differentially expressed mRNAs [DEmRNAs, responding to differentially expressed genes (DEGs)] of microarrays with R package ‘limma’ and the prediction for interactions in the online database. First, DElncRNAs were identified between IDD and control samples in GSE56081. Next, a highly conservative miRNA family profile on the Mircode database (http://​www.​mircode.​org/​) was used to predict interactions between miRNAs for the above DElncRNAs. To validate and ensure the reliability and accuracy of the predicted data, we obtained another set of DEmiRNAs in IDD samples from GSE116726 and GSE19943 for comparison, with the thresholds of P < 0.05 (two-tailed) and |log2 fold change (FC)| ≥ 1. Then, the overlapped miRNAs were used for the final DElncRNA screening.

Prediction of DEmRNAs

As shown in Fig. 2, DEmiRNA targets were predicted using three independent experimental databases, including miRDB (http://​mirdb.​org/​), miRTarBase (http://​mirtarbase.​mbc.​nctu.​edu.​tw/​php/​index.​php), and TargetScan (http://​www.​targetscan.​org/​vert_​72/​). Here, a scoring system was used to predict genes in the DEmRNAs group (DEmRNAs-1). One score was added to the target (mRNA) for each above database that could interact with each other. If the score was ≥2, the mRNA was put into the DEmRNAs-1 group. Meanwhile, the DEmRNAs-2 group was directly identified in GSE56081. Besides, the targets of the overlapped miRNAs from datasets GSE116726 and GSE19943 were predicted as the DEmRNAs-3 group. Next, we kept the overlapped mRNAs in all DEmRNA groups (DEmRNAs-1, DEmRNAs-2, and DEmRNAs-3) as the final DEmRNAs in this study.
After the DE analysis process (Fig. 1), we visualized DElncRNAs, DEmiRNAs, and DEGs between IDD and control samples using clustering heat maps and volcano maps, respectively, using the R package ‘pheatmap’. The key DElncRNAs, DEmiRNAs, and DEGs were selected as the sources of the ceRNA network, which was constructed using Cytoscape3.8.0 [27]. Dark blue rectangle nodes represent the lncRNAs in the network, the purple rhombus nodes represent the target genes, and lines indicate interactions.

Functional enrichment analyses of the ceRNA network

We performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of DEGs using R packages (“clusterProfiler”, “org. Hs.eg.db”, “enrichplot”, and “ggplot2” package) with identified filters of p-Value < 0.05 and q-Value < 0.05 [2830].

Feature selection based on SVM and metabolic gene set

Support vector machine (SVM), a machine learning technology, is a supervised learning model with associated learning algorithms that analyzes data used for classification and regression analyses. Based on the expression levels of the ceRNA, we used the R package ‘caret’ and adopted 5-fold cross-validation to conduct the SVM model for screening vital RNAs. The random seed was set to 39 in all SVM progress. Moreover, we searched for genes on the metabolic gene set based on metabolism-related pathways, which were downloaded from the gene set enrichment analysis (GSEA) website (GSEA, c2.cp.kegg.v7.1.symbols.gmt; https://​www.​gsea-msigdb.​org/​gsea/​downloads.​jsp). We obtained 948 metabolism-related genes for follow-up analysis (Supplementary Table 3). Finally, we used the Venn method (http://​bioinformatics.​psb.​ugent.​be/​webtools/​Venn/​) to search for the key DEG(s) from the results of SVM, expression levels, and the metabolic gene set. Furthermore, we searched for other differentially expressed RNAs (DERNAs) based on the SVM and expression levels to find the critical ceRNAs regulated by the key metabolic gene(s).

Statistical analyses

The R software (version 3.6.2, The R Foundation for Statistical Computing, Vienna, Austria) and MedCalc statistical software (Version 19.0.4, MedCalc Software bvba, Ostend, Belgium) were used for the statistical analyses. Prism (version 8.0, GraphPad Prism Inc.) and R software were used for graphics. DElncRNAs, DEmiRNAs, and DEmRNAs between IDD and control samples were identified using filters with P < 0.05 (two-tailed) and |log2FC| ≥ 1. A positive log2FC value indicated upregulated expression, whereas a negative log2FC value indicated downregulated expression. The chi-square test was used for enrichment analyses. Difference with P < 0.05 were considered statistically significant.

Results

Identification of DElncRNAs, DEmiRNAs, and DEGs using integrated microarray analysis

A total of 502 DElncRNAs and 2295 DEmRNAs were directly identified on GSE56081. Moreover, 15 DEmiRNAs were identified in GSE116726 and GSE19943 (Figs. 1 and 3).

Target prediction for DElncRNAs, DEmiRNA, and DEGs

Of the 502 DElncRNAs, 137 could bind to 207 miRNAs. Target prediction for the above 207 miRNAs revealed that 55 miRNAs could predict the downstream target mRNAs (DEmRNAs-1 group). Surprisingly, we found that miR-338-3p was the only overlapping miRNA between the 55 miRNAs and the 15 DEmiRNAs. Thus, focusing on miR-338-3p, we mined 36 upstream lncRNAs that could bind to it. Furthermore, 366 downstream mRNAs (DEmRNAs-3 group) related to miR-338-3p were also predicted. There were 8612 target mRNAs (DEmRNAs-1 group) of the 55 miRNAs, which were predicted using the miRDB, miRTarBase, and TargetScan databases (Fig. 2). Finally, 45 DEmRNAs (final DEmRNAs, Supplementary Table 4), which bind to miR-338-3p, were the overlapping genes among the 366 mRNAs, 8612 mRNAs, and 2295mRNAs. The 45 DEmRNAs, which combine with miR-338-3p, were concentrated.
To reveal the underlying molecular signatures of IDD, we constructed a ceRNA network using overlapped DElncRNAs and DEGs, the core of which was miR-338-3p. The ceRNA network displayed 45 miR-338-3p/lncRNA links and 36 miR-338-3p/mRNA links (Fig. 4a).

Functional enrichment analyses

The GO terms (45 DEGs) indicated that several genes, which existed in the cell trailing edge, were enriched in the phosphatidylinositol 3-kinase signaling pathway and the cellular response to reactive oxidative stress in biological process (Fig. 4b, details of actual gene IDs and GO descriptions are shown in Supplementary Table 5). Moreover, the KEGG pathway enrichment analysis showed that the 45 DEGs were predominantly enriched in 16 pathways, including the MAPK signaling pathway, IL-17 signaling pathway, apoptosis, and TNF signaling pathway, et al. (Fig. 4c) The details of actual gene IDs per pathway are shown in Supplementary Table 6.

Feature selection based on SVM and metabolic gene set

In dataset GSE56081, we defined 45 overlapping DEGs. Moreover, the metabolic gene set consisted of 948 genes. Hence, we used the SVM method to build a classifier and create a decision boundary for the 36 DElncRNAs and 45 DEGs (Fig. 5a, b). SVM was not used for screening DEmiRNAs because miR-338-3p was the only core DEmiRNA. Then, the Venn method was used to search for the key DElncRNAs and DEGs from the results of SVM signature, expression levels, and the metabolic gene set (Fig. 5c, d, e). The core DElncRNA was lncRNA AC063977.6 (Fig. 5c). The core metabolic gene was 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 2 (PFK/FBPase2) after training by SVM and intersecting with the metabolic gene set (Fig. 5e). The expression levels of core RNAs are presented as a histogram (Fig. 5f).

Discussion

The metabolic homeostasis and the construction of the ECM of intervertebral discs mainly depend on its active NP cells, which form the inner core of the intervertebral disc. An imbalance of cellular functions because of metabolic imbalance, nutrient deprivation, dysregulated apoptosis, and some transcellular signaling interrupts homeostasis in NP cells, resulting in IDD [3133]. Furthermore, the metabolic signature represents a broad molecular signature of systemic metabolism and covers multiple cellular metabolic pathways [34]. The metabolic signature and its relevant mechanism are critical for understanding the cellular mechanisms of various diseases. Accordingly, it would be reasonable to assume that NP cell metabolic disorder might be a pathogenic factor of IDD. However, the metabolic investigations and associated ceRNA regulation mechanism of NP cells in IDD remaine inconclusive. The ceRNAs regulate the expression of various genes and play a significant role in the pathogenesis of many diseases [35]. Identifying ceRNA regulation with the metabolic signatures in NP cells would shed a novel light on IDD. Thus, based on the GEO dataset, we identified a novel gene metabolic signature and related ceRNA regulatory mechanisms to gain insight into the IDD pathogenic mechanism.
In the present study, 45 DEGs and 36 DElncRNAs were identified. MiR-338-3p was the only key DEmiRNA, which was upregulated in IDD. A vast ceRNA network was constructed to reveal the underlying molecular signatures of IDD. GO annotation revealed that the DEmRNAs/DEGs were mostly enriched in the PI3K-Akt signaling pathway. The PI3K-Akt signaling pathway participates in the synthesis of ECM, regulating apoptosis and cell proliferation in IDD. Furthermore, the GO annotation showed that many biological processes were categorized in response to oxidative stress of NP cells, which indicates that oxygen and oxygen-associated processes might play an essential role in IDD. The KEGG pathway enrichment analysis showed that the MAPK signaling pathway, IL-17 signaling pathway, apoptosis, and TNF signaling pathway might be related to IDD. Based on batch normalization, we found that miR-338-3p was the only DEmiRNA validated in two datasets and was correctly predicted for lncRNA-miRNA and mRNA-miRNA binding. However, there were 45 overlapping DEGs and 948 genes defined by the metabolic gene set. To further determine the key metabolic signature and critical lncRNA(s) of the 36 DElncRNAs in IDD, we used the SVM method. Thus, AC063977.6 was identified as the critical lncRNA with recursive feature elimination, based on the results of SVM and related expression regulation of DElncRNAs. Moreover, PFKFB2 is the key metabolic gene with recursive feature elimination, based on the results of SVM, metabolic gene set, and related expression regulation of DEmRNAs.
According to the online database prediction, AC063977.6 directly targets miR-338-3p, whereas miR-338-3p directly targets PFKFB2, suggesting that AC063977.6, miR-338-3p, and PFKFB2 might form an axis that modulates NP cell metabolism in IDD. Furthermore, the gene expression validation based on microarray data of GSE116726 and GSE19943 further enhanced the credibility of this axis. In conclusion, we found that AC063977.6, miR-338-3p, and PFKFB2 formed an axis in IDD to modulate the metabolism of NP cells. This study revealed that miR-338-3p was upregulated in degenerating NP cells, accompanied by downregulation of PFKFB2 expression. Moreover, AC063977.6, acting as a ceRNA sponging miR-338-3p, was significantly attenuated in IDD progression.
The central part of the DEGs is PFKFB2, which is an isoform of the PFKFB family that mainly regulates glycolytic metabolism and catalyzes the fructose 2,6-bisphosphate (Fru-2,6-P) [36]. PFKFB2 is a key bifunctional enzyme involved in glycolytic metabolism [37]. Moreover, glycolysis significantly diminishes cellular oxidative stress [38, 39]. Similarly, bioinformatics analysis with the GEO microarray datasets revealed that the DEGs were significantly enriched in the oxidative stress process in GO enrichment. Compared to normal cells, several cancer cells exhibit elevated expression levels of PFKFB [40]. However, in this study, we found that the expression level of PFKFB2 in NP cells with IDD was lower than that in control NP cells, suggesting that reduced glycolysis may increase the level of cellular oxidative stress in NP cells; thus, it may be associated with IDD.
Accumulating evidence suggests that lncRNAs and miRNAs dysregulate essential pathological processes of IDD, such as apoptosis, angiogenesis, ECM degradation, and inflammatory responses [41]. The miR-338-3p originates from the apoptosis-associated tyrosine kinase gene, and it is dysregulated in many tumors and plays distinct roles in different diseases [42]. The upregulation of miR-338-3p could promote glioma cell invasion and metastasis of lung cancer, whereas the downregulation of miR-338-3p was related to poor outcomes in gastric cancer [4345]. In this study, miR-338-3p was remarkably upregulated in IDD, serving as an impetus with metabolic dysregulation by targeting PFKFB2, thereby disturbing the metabolism of NP cells. IDD severely threatens the health of patients, and identification of genetic signature is beneficial for its early diagnosis and early treatment [23]. Moreover, a strong negative correlation was observed between IDD and PFKFB2 expression levels in both the training cohort and validation cohort, which indicates that low expression of PFKFB2 is a key factor in IDD. The results suggest that PFKFB2 is an player in IDD. According to our experience in clinical work and reported literature, misdiagnosis of IDD occasionally occurs, which often leads to incorrect treatment and even unnecessary surgery. Based on our results, the metabolic signature could be used in providing clues for IDD mechanism research, and may aid in surgery for differential diagnosis in clinical practice.

Limitations of this study

There are some limitations to this study. First, we focused only on the regulatory roles of target genes without further classification into specific subgroups according to their functions, which limited the excavation of those data. Second, further in vitro and in vivo experiments on the sophisticated regulating mechanism involved with AC063977.6 and miR-338 in IDD progression are urgently needed. Third, although a small sample size is a common problem in orthopedic research, it is undeniable that this study is based on a small sample size (a total of 46 samples were included). Sufficient in vitro and in vivo experiments are warranted to establish the functions and mechanisms of the lncRNAs and miRNAs involved in the pathogenesis of IDD and may further promote the translation of precise gene diagnosis in IDD.

Conclusion

The results suggest that lncRNA AC063977.6, acting as a ceRNA that sponges miR-338, correlating with the metabolism of NP cells via the AC063977.6/miR-338/PFKFB2 axis in IDD. PFKFB2 can serve as a metabolic biomarker to contribute to the differential diagnosis of IDD in clinical practice.

Acknowledgments

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare that they do not have anything to disclose regarding conflict of interest with respect to this manuscript.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Risbud MV, Shapiro IM. Role of cytokines in intervertebral disc degeneration: pain and disc content. Nat Rev Rheumatol. 2014;10(1):44–56.CrossRef Risbud MV, Shapiro IM. Role of cytokines in intervertebral disc degeneration: pain and disc content. Nat Rev Rheumatol. 2014;10(1):44–56.CrossRef
2.
Zurück zum Zitat Capoor MN, Ruzicka F, Machackova T, Jancalek R, Smrcka M, Schmitz JE, Hermanova M, Sana J, Michu E, Baird JC, et al. Prevalence of Propionibacterium acnes in intervertebral discs of patients undergoing lumbar microdiscectomy: a prospective cross-sectional study. PLoS One. 2016;11(8):e0161676.CrossRef Capoor MN, Ruzicka F, Machackova T, Jancalek R, Smrcka M, Schmitz JE, Hermanova M, Sana J, Michu E, Baird JC, et al. Prevalence of Propionibacterium acnes in intervertebral discs of patients undergoing lumbar microdiscectomy: a prospective cross-sectional study. PLoS One. 2016;11(8):e0161676.CrossRef
3.
Zurück zum Zitat Zhao K, Zhang Y, Yuan H, Zhao M, Zhao D. Long noncoding RNA LINC00958 accelerates the proliferation and matrix degradation of the nucleus pulposus by regulating miR-203/SMAD3. Aging. 2019;11(23):10814–25.CrossRef Zhao K, Zhang Y, Yuan H, Zhao M, Zhao D. Long noncoding RNA LINC00958 accelerates the proliferation and matrix degradation of the nucleus pulposus by regulating miR-203/SMAD3. Aging. 2019;11(23):10814–25.CrossRef
4.
Zurück zum Zitat Jensen M, Röthlisberger U, Rovira C. Hydroxide and proton migration in aquaporins. Biophys J. 2005;89(3):1744–59.CrossRef Jensen M, Röthlisberger U, Rovira C. Hydroxide and proton migration in aquaporins. Biophys J. 2005;89(3):1744–59.CrossRef
5.
Zurück zum Zitat Leveillard T, Philp NJ, Sennlaub F. Is Retinal Metabolic Dysfunction at the Center of the Pathogenesis of Age-related Macular Degeneration? Int J Mol Sci. 2019;20(3). Leveillard T, Philp NJ, Sennlaub F. Is Retinal Metabolic Dysfunction at the Center of the Pathogenesis of Age-related Macular Degeneration? Int J Mol Sci. 2019;20(3).
6.
Zurück zum Zitat Allegra A, Innao V, Gerace D, Bianco O, Musolino C. The metabolomic signature of hematologic malignancies. Leuk Res. 2016;49:22–35.CrossRef Allegra A, Innao V, Gerace D, Bianco O, Musolino C. The metabolomic signature of hematologic malignancies. Leuk Res. 2016;49:22–35.CrossRef
7.
Zurück zum Zitat Chang ML, Yang SS. Metabolic Signature of Hepatic Fibrosis: From Individual Pathways to Systems Biology. Cells. 2019;8(11). Chang ML, Yang SS. Metabolic Signature of Hepatic Fibrosis: From Individual Pathways to Systems Biology. Cells. 2019;8(11).
8.
Zurück zum Zitat Dumas ME, Davidovic L. Metabolic profiling and Phenotyping of central nervous system diseases: metabolites bring insights into brain dysfunctions. J NeuroImmune Pharmacol. 2015;10(3):402–24.CrossRef Dumas ME, Davidovic L. Metabolic profiling and Phenotyping of central nervous system diseases: metabolites bring insights into brain dysfunctions. J NeuroImmune Pharmacol. 2015;10(3):402–24.CrossRef
9.
Zurück zum Zitat Li P, Xue WJ, Feng Y, Mao QS. Long non-coding RNA CASC2 suppresses the proliferation of gastric cancer cells by regulating the MAPK signaling pathway. Am J Transl Res. 2016;8(8):3522–9.PubMedPubMedCentral Li P, Xue WJ, Feng Y, Mao QS. Long non-coding RNA CASC2 suppresses the proliferation of gastric cancer cells by regulating the MAPK signaling pathway. Am J Transl Res. 2016;8(8):3522–9.PubMedPubMedCentral
10.
Zurück zum Zitat Zhao W, Geng D, Li S, Chen Z, Sun M. LncRNA HOTAIR influences cell growth, migration, invasion, and apoptosis via the miR-20a-5p/HMGA2 axis in breast cancer. Cancer Med. 2018;7(3):842–55.CrossRef Zhao W, Geng D, Li S, Chen Z, Sun M. LncRNA HOTAIR influences cell growth, migration, invasion, and apoptosis via the miR-20a-5p/HMGA2 axis in breast cancer. Cancer Med. 2018;7(3):842–55.CrossRef
11.
Zurück zum Zitat Zhan S, Wang K, Xiang Q, Song Y, Li S, Liang H, Luo R, Wang B, Liao Z, Zhang Y, et al. lncRNA HOTAIR upregulates autophagy to promote apoptosis and senescence of nucleus pulposus cells. J Cell Physiol. 2020;235(3):2195–208.CrossRef Zhan S, Wang K, Xiang Q, Song Y, Li S, Liang H, Luo R, Wang B, Liao Z, Zhang Y, et al. lncRNA HOTAIR upregulates autophagy to promote apoptosis and senescence of nucleus pulposus cells. J Cell Physiol. 2020;235(3):2195–208.CrossRef
12.
Zurück zum Zitat Ma F, Wang SH, Cai Q, Jin LY, Zhou D, Ding J, Quan ZW. Long non-coding RNA TUG1 promotes cell proliferation and metastasis by negatively regulating miR-300 in gallbladder carcinoma. Biomed Pharmacother. 2017;88:863–9.CrossRef Ma F, Wang SH, Cai Q, Jin LY, Zhou D, Ding J, Quan ZW. Long non-coding RNA TUG1 promotes cell proliferation and metastasis by negatively regulating miR-300 in gallbladder carcinoma. Biomed Pharmacother. 2017;88:863–9.CrossRef
13.
Zurück zum Zitat Tay Y, Rinn J, Pandolfi PP. The multilayered complexity of ceRNA crosstalk and competition. Nature. 2014;505(7483):344–52.CrossRef Tay Y, Rinn J, Pandolfi PP. The multilayered complexity of ceRNA crosstalk and competition. Nature. 2014;505(7483):344–52.CrossRef
14.
Zurück zum Zitat Zhang CZ. Long non-coding RNA FTH1P3 facilitates oral squamous cell carcinoma progression by acting as a molecular sponge of miR-224-5p to modulate fizzled 5 expression. Gene. 2017;607:47–55.CrossRef Zhang CZ. Long non-coding RNA FTH1P3 facilitates oral squamous cell carcinoma progression by acting as a molecular sponge of miR-224-5p to modulate fizzled 5 expression. Gene. 2017;607:47–55.CrossRef
15.
Zurück zum Zitat Smillie CL, Sirey T, Ponting CP. Complexities of post-transcriptional regulation and the modeling of ceRNA crosstalk. Crit Rev Biochem Mol Biol. 2018;53(3):231–45.CrossRef Smillie CL, Sirey T, Ponting CP. Complexities of post-transcriptional regulation and the modeling of ceRNA crosstalk. Crit Rev Biochem Mol Biol. 2018;53(3):231–45.CrossRef
16.
Zurück zum Zitat Han Z, Wang J, Gao L, Wang Q, Wu J. Aberrantly expressed messenger RNAs and long noncoding RNAs in degenerative nucleus pulposus cells co-cultured with adipose-derived mesenchymal stem cells. Arthritis Res Ther. 2018;20(1):182.CrossRef Han Z, Wang J, Gao L, Wang Q, Wu J. Aberrantly expressed messenger RNAs and long noncoding RNAs in degenerative nucleus pulposus cells co-cultured with adipose-derived mesenchymal stem cells. Arthritis Res Ther. 2018;20(1):182.CrossRef
17.
Zurück zum Zitat Xi Y, Jiang T, Wang W, Yu J, Wang Y, Wu X, He Y. Long non-coding HCG18 promotes intervertebral disc degeneration by sponging miR-146a-5p and regulating TRAF6 expression. Sci Rep. 2017;7(1):13234.CrossRef Xi Y, Jiang T, Wang W, Yu J, Wang Y, Wu X, He Y. Long non-coding HCG18 promotes intervertebral disc degeneration by sponging miR-146a-5p and regulating TRAF6 expression. Sci Rep. 2017;7(1):13234.CrossRef
18.
Zurück zum Zitat Sanz H, Valim C, Vegas E, Oller JM, Reverter F. SVM-RFE: selection and visualization of the most relevant features through non-linear kernels. BMC Bioinformatics. 2018;19(1):432.CrossRef Sanz H, Valim C, Vegas E, Oller JM, Reverter F. SVM-RFE: selection and visualization of the most relevant features through non-linear kernels. BMC Bioinformatics. 2018;19(1):432.CrossRef
19.
Zurück zum Zitat Xu JX, Yang SD, Wang BL, Yang DL, Ding WY, Shen Y. Correlative analyses of isolated upper lumbar disc herniation and adjacent wedge-shaped vertebrae. Int J Clin Exp Med. 2015;8(1):1150–5.PubMedPubMedCentral Xu JX, Yang SD, Wang BL, Yang DL, Ding WY, Shen Y. Correlative analyses of isolated upper lumbar disc herniation and adjacent wedge-shaped vertebrae. Int J Clin Exp Med. 2015;8(1):1150–5.PubMedPubMedCentral
20.
Zurück zum Zitat Izzo R, Popolizio T, D'Aprile P, Muto M. Spinal pain. Eur J Radiol. 2015;84(5):746–56.CrossRef Izzo R, Popolizio T, D'Aprile P, Muto M. Spinal pain. Eur J Radiol. 2015;84(5):746–56.CrossRef
21.
Zurück zum Zitat Zarrabian MM, Diehn FE, Kotsenas AL, Wald JT, Yu E, Nassr A. Dorsal lumbar disc migrations with lateral and ventral epidural extension on axial MRI: a case series and review of the literature. AJNR Am J Neuroradiol. 2016;37(11):2171–7.CrossRef Zarrabian MM, Diehn FE, Kotsenas AL, Wald JT, Yu E, Nassr A. Dorsal lumbar disc migrations with lateral and ventral epidural extension on axial MRI: a case series and review of the literature. AJNR Am J Neuroradiol. 2016;37(11):2171–7.CrossRef
22.
Zurück zum Zitat Ghaly RF, Perciuleac Z, Candido KD, Knezevic NN. Athletic pubalgia misdiagnosed as lumbar radiculopathy - a case report. Surg Neurol Int. 2019;10:224.CrossRef Ghaly RF, Perciuleac Z, Candido KD, Knezevic NN. Athletic pubalgia misdiagnosed as lumbar radiculopathy - a case report. Surg Neurol Int. 2019;10:224.CrossRef
23.
Zurück zum Zitat Feng Y, Egan B, Wang J. Genetic factors in intervertebral disc degeneration. Genes Dis. 2016;3(3):178–85.CrossRef Feng Y, Egan B, Wang J. Genetic factors in intervertebral disc degeneration. Genes Dis. 2016;3(3):178–85.CrossRef
24.
Zurück zum Zitat Wu ZH, Tang Y, Zhou Y. A Metabolic Gene Signature to Predict Overall Survival in Head and Neck Squamous Cell Carcinoma. Mediat Inflamm. 2020;2020:6716908. Wu ZH, Tang Y, Zhou Y. A Metabolic Gene Signature to Predict Overall Survival in Head and Neck Squamous Cell Carcinoma. Mediat Inflamm. 2020;2020:6716908.
25.
Zurück zum Zitat Zeng F, Su J, Peng C, Liao M, Zhao S, Guo Y, Chen X, Deng G. Prognostic implications of metabolism related gene signature in cutaneous melanoma. Front Oncol. 2020;10:1710.CrossRef Zeng F, Su J, Peng C, Liao M, Zhao S, Guo Y, Chen X, Deng G. Prognostic implications of metabolism related gene signature in cutaneous melanoma. Front Oncol. 2020;10:1710.CrossRef
26.
Zurück zum Zitat Gruber HE, Hoelscher GL, Ingram JA, Hanley EN Jr. Genome-wide analysis of pain-, nerve- and neurotrophin -related gene expression in the degenerating human annulus. Mol Pain. 2012;8:63.CrossRef Gruber HE, Hoelscher GL, Ingram JA, Hanley EN Jr. Genome-wide analysis of pain-, nerve- and neurotrophin -related gene expression in the degenerating human annulus. Mol Pain. 2012;8:63.CrossRef
27.
Zurück zum Zitat Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.CrossRef Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.CrossRef
28.
Zurück zum Zitat Kanehisa M. Toward understanding the origin and evolution of cellular organisms. Protein Sci. 2019;28(11):1947–51.CrossRef Kanehisa M. Toward understanding the origin and evolution of cellular organisms. Protein Sci. 2019;28(11):1947–51.CrossRef
29.
Zurück zum Zitat Kanehisa M, Furumichi M, Sato Y, Ishiguro-Watanabe M, Tanabe M. KEGG: integrating viruses and cellular organisms. Nucleic Acids Res. 2021;49(D1):D545–d551.CrossRef Kanehisa M, Furumichi M, Sato Y, Ishiguro-Watanabe M, Tanabe M. KEGG: integrating viruses and cellular organisms. Nucleic Acids Res. 2021;49(D1):D545–d551.CrossRef
30.
Zurück zum Zitat Kanehisa M, Goto S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27–30.CrossRef Kanehisa M, Goto S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27–30.CrossRef
31.
Zurück zum Zitat Chen D, Xia D, Pan Z, Xu D, Zhou Y, Wu Y, Cai N, Tang Q, Wang C, Yan M, et al. Metformin protects against apoptosis and senescence in nucleus pulposus cells and ameliorates disc degeneration in vivo. Cell Death Dis. 2016;7(10):e2441.CrossRef Chen D, Xia D, Pan Z, Xu D, Zhou Y, Wu Y, Cai N, Tang Q, Wang C, Yan M, et al. Metformin protects against apoptosis and senescence in nucleus pulposus cells and ameliorates disc degeneration in vivo. Cell Death Dis. 2016;7(10):e2441.CrossRef
32.
Zurück zum Zitat Liu J, Yuan C, Pu L, Wang J. Nutrient deprivation induces apoptosis of nucleus pulposus cells via activation of the BNIP3/AIF signalling pathway. Mol Med Rep. 2017;16(5):7253–60.CrossRef Liu J, Yuan C, Pu L, Wang J. Nutrient deprivation induces apoptosis of nucleus pulposus cells via activation of the BNIP3/AIF signalling pathway. Mol Med Rep. 2017;16(5):7253–60.CrossRef
33.
Zurück zum Zitat Liao Z, Luo R, Li G, Song Y, Zhan S, Zhao K, Hua W, Zhang Y, Wu X, Yang C. Exosomes from mesenchymal stem cells modulate endoplasmic reticulum stress to protect against nucleus pulposus cell death and ameliorate intervertebral disc degeneration in vivo. Theranostics. 2019;9(14):4084–100.CrossRef Liao Z, Luo R, Li G, Song Y, Zhan S, Zhao K, Hua W, Zhang Y, Wu X, Yang C. Exosomes from mesenchymal stem cells modulate endoplasmic reticulum stress to protect against nucleus pulposus cell death and ameliorate intervertebral disc degeneration in vivo. Theranostics. 2019;9(14):4084–100.CrossRef
34.
Zurück zum Zitat Park HJ, Kim S, Li W. Model-based analysis of competing-endogenous pathways (MACPath) in human cancers. PLoS Comput Biol. 2018;14(3):e1006074.CrossRef Park HJ, Kim S, Li W. Model-based analysis of competing-endogenous pathways (MACPath) in human cancers. PLoS Comput Biol. 2018;14(3):e1006074.CrossRef
35.
Zurück zum Zitat Yan J, Song J, Qiao M, Zhao X, Li R, Jiao J, Sun Q. Long noncoding RNA expression profile and functional analysis in psoriasis. Mol Med Rep. 2019;19(5):3421–30.PubMedPubMedCentral Yan J, Song J, Qiao M, Zhao X, Li R, Jiao J, Sun Q. Long noncoding RNA expression profile and functional analysis in psoriasis. Mol Med Rep. 2019;19(5):3421–30.PubMedPubMedCentral
36.
Zurück zum Zitat Wang X, Li D, Wu H, Liu F, Liu F, Zhang Q, Li J. LncRNA TRPC7-AS1 regulates nucleus pulposus cellular senescence and ECM synthesis via competing with HPN for miR-4769-5p binding. Mech Ageing Dev. 2020;190:111293. Wang X, Li D, Wu H, Liu F, Liu F, Zhang Q, Li J. LncRNA TRPC7-AS1 regulates nucleus pulposus cellular senescence and ECM synthesis via competing with HPN for miR-4769-5p binding. Mech Ageing Dev. 2020;190:111293.
37.
Zurück zum Zitat Barik S. An intronic microRNA silences genes that are functionally antagonistic to its host gene. Nucleic Acids Res. 2008;36(16):5232–41. Barik S. An intronic microRNA silences genes that are functionally antagonistic to its host gene. Nucleic Acids Res. 2008;36(16):5232–41.
38.
Zurück zum Zitat Chen X, Wei L, Zhao S. miR-338 inhibits the metastasis of lung cancer by targeting integrin beta3. Oncol Rep. 2016;36(3):1467–74. Chen X, Wei L, Zhao S. miR-338 inhibits the metastasis of lung cancer by targeting integrin beta3. Oncol Rep. 2016;36(3):1467–74.
39.
Zurück zum Zitat Li Y, Huang Y, Qi Z, Sun T, Zhou Y. MiR-338-5p promotes Glioma cell invasion by regulating TSHZ3 and MMP2. Cell Mol Neurobiol. 2018;38(3):669–77. Li Y, Huang Y, Qi Z, Sun T, Zhou Y. MiR-338-5p promotes Glioma cell invasion by regulating TSHZ3 and MMP2. Cell Mol Neurobiol. 2018;38(3):669–77.
40.
Zurück zum Zitat Liu S, Suo J, Wang C, Sun X, Wang D, He L, Zhang Y, Li W. Downregulation of tissue miR-338-3p predicts unfavorable prognosis of gastric cancer. Cancer Biomark. 2017;21(1):117–22. Liu S, Suo J, Wang C, Sun X, Wang D, He L, Zhang Y, Li W. Downregulation of tissue miR-338-3p predicts unfavorable prognosis of gastric cancer. Cancer Biomark. 2017;21(1):117–22.
41.
Zurück zum Zitat Arden C, Hampson LJ, Huang GC, Shaw JA, Aldibbiat A, Holliman G, Manas D, Khan S, Lange AJ, Agius L. A role for PFK-2/FBPase-2, as distinct from fructose 2,6-bisphosphate, in regulation of insulin secretion in pancreatic beta-cells. Biochem J. 2008;411(1):41–51. Arden C, Hampson LJ, Huang GC, Shaw JA, Aldibbiat A, Holliman G, Manas D, Khan S, Lange AJ, Agius L. A role for PFK-2/FBPase-2, as distinct from fructose 2,6-bisphosphate, in regulation of insulin secretion in pancreatic beta-cells. Biochem J. 2008;411(1):41–51.
42.
Zurück zum Zitat Schmidt S, Rainer J, Riml S, Ploner C, Jesacher S, Achmuller C, Presul E, Skvortsov S, Crazzolara R, Fiegl M, et al. Identification of glucocorticoid-response genes in children with acute lymphoblastic leukemia. Blood. 2006;107(5):2061–9. Schmidt S, Rainer J, Riml S, Ploner C, Jesacher S, Achmuller C, Presul E, Skvortsov S, Crazzolara R, Fiegl M, et al. Identification of glucocorticoid-response genes in children with acute lymphoblastic leukemia. Blood. 2006;107(5):2061–9.
43.
Zurück zum Zitat Brand KA, Hermfisse U. Aerobic glycolysis by proliferating cells: a protective strategy against reactive oxygen species. FASEB J. 1997;11(5):388–95. Brand KA, Hermfisse U. Aerobic glycolysis by proliferating cells: a protective strategy against reactive oxygen species. FASEB J. 1997;11(5):388–95.
44.
Zurück zum Zitat Poljsak B, Kovac V, Dahmane R, Levec T, Starc A. Cancer etiology: a metabolic disease originating from Life's major evolutionary transition? Oxidative Med Cell Longev. 2019;2019:7831952. Poljsak B, Kovac V, Dahmane R, Levec T, Starc A. Cancer etiology: a metabolic disease originating from Life's major evolutionary transition? Oxidative Med Cell Longev. 2019;2019:7831952.
45.
Zurück zum Zitat Pegoraro C, Maczkowiak F, Monsoro-Burq AH. Pfkfb (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase) isoforms display a tissue-specific and dynamic expression during Xenopus laevis development. Gene Expr Patterns. 2013;13(7):203–11. Pegoraro C, Maczkowiak F, Monsoro-Burq AH. Pfkfb (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase) isoforms display a tissue-specific and dynamic expression during Xenopus laevis development. Gene Expr Patterns. 2013;13(7):203–11.
Metadaten
Titel
Major ceRNA regulation and key metabolic signature analysis of intervertebral disc degeneration
verfasst von
Shuai Cao
Jie Li
Kai Yang
Haopeng Li
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
BMC Musculoskeletal Disorders / Ausgabe 1/2021
Elektronische ISSN: 1471-2474
DOI
https://doi.org/10.1186/s12891-021-04109-8

Weitere Artikel der Ausgabe 1/2021

BMC Musculoskeletal Disorders 1/2021 Zur Ausgabe

Arthropedia

Grundlagenwissen der Arthroskopie und Gelenkchirurgie. Erweitert durch Fallbeispiele, Videos und Abbildungen. 
» Jetzt entdecken

Update Orthopädie und Unfallchirurgie

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