#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Genome-wide analysis of DNA methylation profile identifies differentially methylated loci associated with human intervertebral disc degeneration


Authors: Akihiro Ikuno aff001;  Koji Akeda aff002;  Shin-ichiro Takebayashi aff001;  Motomu Shimaoka aff003;  Katsuzumi Okumura aff001;  Akihiro Sudo aff002
Authors place of work: Laboratory of Molecular & Cellular Biology, Graduate School of Bioresources, Mie University, Tsu, Japan aff001;  Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu, Japan aff002;  Department of Molecular Pathobiology3, Mie University Graduate School of Medicine, Tsu, Japan aff003
Published in the journal: PLoS ONE 14(9)
Category: Research Article
doi: https://doi.org/10.1371/journal.pone.0222188

Summary

Background

Environmental and endogenous factors under genetic predisposition are considered to initiate the human intervertebral disc (IVD) degeneration. DNA methylation is an essential mechanism to ensure cell-specific gene expression for normal development and tissue stability. Aberrant epigenetic alterations play a pivotal role in several diseases, including osteoarthritis. However, epigenetic alternations, including DNA methylation, in IVD degeneration have not been evaluated. The purpose of this study was to comprehensively compare the genome-wide DNA methylation profiles of human IVD tissues, specifically nucleus pulpous (NP) tissues, with early and advanced stages of disc degeneration.

Methods

Human NP tissues were used in this study. The samples were divided into two groups: early stage degeneration (n = 8, Pfirrmann’s MRI grade: I-III) and advanced stage degeneration (n = 8, grade: IV). Genomic DNA was processed for genome-wide DNA methylation profiling using the Infinium MethylationEPIC BeadChip array. Extraction of raw methylation data, clustering and scatter plot of each group values of each sample were performed using a methylation module in GenomeStudio software. The identification of differentially methylated loci (DMLs) and the Gene Ontology (GO) analysis were performed using R software with the ChAMP package.

Results

Unsupervised hierarchical clustering revealed that early and advanced stage degenerated IVD samples segregated into two main clusters by their DNA methylome. A total of 220 DMLs were identified between early and advanced disc degeneration stages. Among these, four loci were hypomethylated and 216 loci were hypermethylated in the advanced disc degeneration stage. The GO enrichment analysis of genes containing DMLs identified two significant GO terms for biological processes, hemophilic cell adhesion and cell-cell adhesion.

Conclusions

We conducted a genome-wide DNA methylation profile comparative study and observed significant differences in DNA methylation profiles between early and advanced stages of human IVD degeneration. These results implicate DNA methylation in the process of human IVD degeneration.

Keywords:

DNA – Biology and life sciences – Cell biology – Chromosome biology – Chromatin – Chromatin modification – DNA methylation – Genetics – Epigenetics – DNA modification – Gene expression – Genomics – Genome analysis – Biochemistry – Nucleic acids – Computational biology – Research and analysis methods – Genetic loci – Medicine and health sciences – Cellular structures and organelles – Diagnostic medicine – Rheumatology – arthritis – Imaging techniques – Diagnostic radiology – magnetic resonance imaging – Radiology and imaging – signal transduction – Cell signaling – Signaling cascades – Wnt signaling cascade – Hedgehog signaling – extracellular matrix – Gene ontologies – osteoarthritis

Introduction

Low back pain (LBP) is a debilitating disorder that is significantly associated with personal, social, and economic burdens. Recent reports in the Global Burden of Disease (GBD) Study 2015 showed that 7.3% of the global population (540 million people) had activity-limiting LBP on the global point prevalence survey[1].

Epidemiological and clinical studies have recently provided evidence that LBP has a significant association with lumbar intervertebral disc (IVD) degeneration[26].

The vertebral column complex consists of ventrally located vertebral bodies and intervening intervertebral discs (IVDs). The IVD is composed of a central gelatinous nucleus pulposus (NP) and a surrounding fibrous anulus fibrosus (AF).

Intervertebral disc degeneration is suggested to be defined as ‘the structural and functional failure of the disc as a result of aberrant, pathological cellular and extracellular matrix (ECM) changes'[7]. The pathophysiology of IVD degeneration is not entirely understood; however, environmental and endogenous factors under genetic predisposition are considered to initiate the degenerative changes of human IVDs (see review in[8]). Intervertebral disc degeneration is generally believed to be a consequence of increased catabolism of the ECM[8, 9]. Biochemically, IVD degeneration, especially NP degeneration, is well characterized by a change in extracellular matrix molecules (loss of proteoglycan and water content in the NP), resulting in an alteration of the biomechanical properties of IVD tissues. These degenerative changes are considered to induce the disruption of IVD tissues, leading to the degenerative disc diseases that are associated with low back pain[9].

A substantial number of mechanisms are known that regulate gene expression and cell fate persistence, commonly referred to as epigenetics[10]. The most extensively studied epigenetic modulation is DNA methylation[11].

DNA methylation induces changes in gene expression without changing the DNA sequence by adding methyl groups to a cytosine in a CpG-containing nucleotide to form 5-methylcytosine[12]. When methylation is located in gene promoter and enhancer regions, DNA methylation typically acts to silence genes, whereas methylation located in gene body regions usually induce enhanced gene expression[13]. DNA methylation is an essential mechanism to ensure cell-type-specific gene expression for normal development, while aberrant epigenetic alterations have been considered to play a pivotal role in several different diseases, such as cancer and neurodegenerative diseases[14, 15]. Therefore, research in epigenetics, including DNA methylation, can elucidate the key pathological process of several diseases; hence, leading to the identification of a new molecular target for therapeutic intervention.

Osteoarthritis (OA) is a chronic musculoskeletal disease characterized by degradation of articular cartilage; similar biochemical changes have also been found in the pathogenesis of IVD degeneration. The involvement of DNA methylation in the pathogenesis of OA has been increasingly evident, reflected by the growing body of reports on the subject. Cross-Sectional studies of DNA methylation on candidate genes have identified alternations in the methylation status of genes involved in OA pathogenesis[1623]. More recently, genome-wide DNA methylation studies have shown that there is a distinct methylation profile in OA cartilage compared with healthy cartilage in the hip and knee joints[2431]. However, epigenetic alternations, including DNA methylation, in IVD degeneration have not been evaluated.

The purpose of this study was to comprehensively compare the Genome-wide DNA methylation profiles of human NP tissues at early and advanced stages of disc degeneration using an Infinium MethylationEPIC BeadChip array.

Materials and methods

Human intervertebral disc samples

Study ethics were approved by the institutional review board of the Mie University Hospital (Tsu, Mie, Japan; IRB reference number: H2018-050). Written or oral informed consent was obtained from all participants.

Human IVD tissues obtained from spine surgeries were used in this study (average age: 55.6 [2583] years-old). The degree of disc degeneration was evaluated by preoperative magnetic resonance imaging (MRI) according to Pfirrmann’s classification[32]: grade I (n = 3); grade II (n = 3); grade III (n = 2); grade IV (n = 8) (Fig 1). Human IVD tissues were divided into two groups: early stage degeneration (Pfirrmann grades I-III, Fig 1) and advanced stage degeneration (Pfirrmann grades IV–V, Fig 1) (Table 1). NP tissues grossly separated from human IVD samples were stored at -80°C until used.

<h2>Magnetic resonance imaging (MRI) of human intervertebral disc classified according to the Pfirrmann grading system[<em class="ref">22</em>].</h2>
Fig. 1.

Magnetic resonance imaging (MRI) of human intervertebral disc classified according to the Pfirrmann grading system[22].


Grades I to III were classified as early stage degeneration (ED). Grade IV and V were classified as advanced stage degeneration (AD).

Tab. 1.

Patient characteristics.

<h2>Patient characteristics.</h2>

DNA isolation and bisulfate treatment

Frozen NP tissue samples (200–250 mg wet weight) were pulverized in the presence of liquid nitrogen using a cryopress (Microtech Nichion, Chiba, Japan). DNA was isolated using the Wizard® Genomic DNA Purification Kit (Promega, City, WI, USA) according to the manufacturer's instruction. First, protein precipitation solution, 0.5 M EDTA and nuclei lysis solution were added to the sample (20 mg), and the sample was then treated with Proteinase K. Next, DNA was precipitated by adding isopropanol. Finally, the DNA pellet was washed twice in 70% ethanol and resuspended in sterilized ultrapure water. The concentration of DNA was measured by the Qubit® dsDNA HS Assay Kit (Molecular Probes, City, OR, USA). The DNA samples were stored at -20°C. Five hundred ng of genomic DNA was then bisulfite converted using an EZ DNA methylation kit (Zymo Research, Irvine, CA, USA) and eluted in 10 μl of elution buffer (50 ng/μl).

Genome-wide DNA methylation profiling

DNA methylation profiling was performed on bisulfite-converted genomic DNA in the Center for Molecular Biology and Genetics of Mie University using the Infinium MethylationEPIC BeadChip array, which allowed the interrogation of over 850,000 methylation sites throughout the genome at single-nucleotide resolution (Line #000010, catalog #WG-317-1001, Illumina, San Diego, CA, USA). The arrays were processed following the manufacturer's instructions and scanned in an Illumina iScan (Illumina). Extraction of raw methylation data, scatter plots of each group values and clustering of each sample were performed using the Methylation module (Version 1.90) in GenomeStudio software (V2011.1, Illumina). GenomeStudio provides the methylation data as β values: β = M/(M + U), which were calculated from the fluorescent signal of the methylation probe (M) and unmethylated probe (U). The β values range from 0 (no methylation) to 1 (100% methylation).

A difference in β values between early and advanced IVD degeneration stage groups were tested with the Illumina Custom model. False discovery rate (FDR)—corrected P values and DiffScores were computed. The data of the Infinium MethylationEPIC BeadChip array are available on NCBI NIH Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo/) under accession number # GSE129789. For scatter plot and clustering analyses, raw data were normalized and background was subtracted using a control probe in this array. Probes that had a detection P—value greater than 0.01 were removed from the analysis data. Because male and female samples were studied, sex chromosome probes were also removed.

Data processing and statistical analysis

Processing of the raw methylation data was performed using R (version 3.4.3; https://www.r-project.org/) with the Chip Analysis Methylation Pipeline (ChAMP) package[33]. Raw methylation data were imported by the minfi method[34, 35], and normalized by the SWAN method[36]. By default setting, raw data were filtered for probes with a detection P > 0.01, non-CpG site[37], the multi-hit probe list[38] or X and Y chromosomes. As a result, the remaining 741955 probes were utilized for data analysis. To show statistically significant genome-wide differences in the differential methylated loci (DMLs), the adjustment P-value (Adjust P.Value) moderated with “BH (Benjamini-Hochberg)” correction was calculated using the limma package[39, 40]. DMLs whose Adjust P.Value was less than 0.05 were selected. Gene ontology analysis for 220 DMLs was performed using the missMethyl package with the gometh method[4143].

Comparison analysis of differentially methylated loci (DMLs) with human knee osteoarthritis

To identify the common gene symbols that differentially methylated between human IVD degeneration and human knee osteoarthritis, 187 gene symbols comprising 220 DMLs identified in this study were compared with 484 gene symbols comprising 653 DMLs identified in knee osteoarthritis (OA) cartilage study[44]. The percentage of common gene symbols against 187 genes and the percentage of the DMLs associated with common gene symbols against 220 DMLs were calculated.

Statistical analysis

The correlation between methylation β values and age was evaluated using Pearson’s correlation coefficient test. Differences in methylation β values were assessed for statistical significance by two-way analysis of variance (ANOVA) to compare the disc degeneration groups (ED and AD) and gender. All the statistical analyses were performed using IBM Statistical Package for Social Sciences Software (SPSS) Statistics (IBM Japan, Tokyo). The accepted level of significance was p<0.05.

Results

DNA methylome in early and advanced stages of human intervertebral disc degeneration

Unsupervised hierarchical clustering revealed that early and advanced stages of degenerated samples segregated into two main clusters by their DNA methylome (Fig 2). Cluster 1 consists of 7 ED samples and 3 AD samples and cluster 2 consists of 5 AD samples and 1 ED sample. Scatter plot of average methylation β values in all ED and AD samples are presented in Fig 3.

<h2>Unsupervised hierarchical clustering of DNA methylation values of human nucleus pulposus (NP) tissues in eight early stage (ED) and eight advanced stage (AD) of disc degeneration.</h2>
Fig. 2.

Unsupervised hierarchical clustering of DNA methylation values of human nucleus pulposus (NP) tissues in eight early stage (ED) and eight advanced stage (AD) of disc degeneration.


<h2>Scatter plot of average DNA methylation level of early stage (ED) and advanced stage (AD) of degeneration.</h2>
Fig. 3.

Scatter plot of average DNA methylation level of early stage (ED) and advanced stage (AD) of degeneration.


Identification of differentially methylated loci (DMLs) in early and advanced stages of human disc degeneration

A total of 220 differentially methylated loci (DMLs) were identified in early and advanced IVD degeneration stages, comprising a total of 187 individual genes (for the complete list of DMLs, see supporting information, S1 Table). Among these, four loci were hypomethylated, and 216 sites were hypermethylated in the advanced stage of degenerated IVDs. The gene-associated four hypomethylated DMLs and ten highest hypermethylated DMLs in the advanced IVD degeneration stage are shown in Table 2.

Tab. 2.

Analysis of significantly differentially methylated loci (DMLs).

<h2>Analysis of significantly differentially methylated loci (DMLs).</h2>

Examples of methylation β value plots for the four representative hypomethylated and hypermethylated DMLs are shown in Fig 4. In the hypomethylated DMLs, the averaged β value of CARD14, CRHR1, C14orf139 and ZBTB47 in the ED group was significantly higher compared to those in the AD group (Fig 4A–4D). In the hypermethylated DMLs, the averaged β value of GNL3, SNORA52, XYR5 and MED23 in the ED group was significantly lower compared to those in the AD group (Fig 4E–4H).

<h2>Differences in β value of four highest hypomethylated and hypermethylated loci between early stage (ED) and advanced stage (AD) of degeneration.</h2>
Fig. 4.

Differences in β value of four highest hypomethylated and hypermethylated loci between early stage (ED) and advanced stage (AD) of degeneration.


* = adjust. P. Val < 0.05; ** = adjust. P. Val < 0.01; *** = adjust. P. Val < 0.001. A: CARD14 (Caspase Recruitment Domain Family Member 14), B: CRHR1 (Corticotropin Releasing Hormone Receptor 1), C: C14orf139 (Chromosome 14 Open Reading Frame 139), D: ZBTB47 (Zinc Finger And BTB Domain Containing 47), E: GNL3 (G Protein Nucleolar 3), F: SNORA52 (Small Nucleolar RNA, H/ACA Box 52), G: XKR5 (X Kell Blood Group Precursor-Related Family, Member 5), H: MED23 (Mediator Complex Subunit 23).

In the total eight samples including ED and AD groups, a significant correlation between methylation β-value and age was found in 6 genes (CARD14, CRHR1, GNL3, SNORA52, XKR5, and MED23); however, the remaining two genes (C24orf139 and ZBTB47) showed no significant correlation between methylation β value and age (Fig 5). When the data were analyzed by ED and AD groups, respectively, no significant correlation between methylation β values and age were identified both by ED and AD groups (Fig 5).

<h2>Scatter plots of the β value of four highest hypomethylated and hypermethylated loci between early stage (ED) and advanced stage (AD) of degeneration according to age.</h2>
Fig. 5.

Scatter plots of the β value of four highest hypomethylated and hypermethylated loci between early stage (ED) and advanced stage (AD) of degeneration according to age.


Blue dots indicate the β values of the early stage of degenerated (ED) samples, and orange dots indicate β values of the advanced stage of degenerated (AD) samples. R2: correlation coefficient. *P<0.05, **P<0.01. A: CARD14 (Caspase Recruitment Domain Family Member 14), B: CRHR1 (Corticotropin Releasing Hormone Receptor 1), C: C14orf139 (Chromosome 14 Open Reading Frame 139), D: ZBTB47 (Zinc Finger And BTB Domain Containing 47), E: GNL3 (G Protein Nucleolar 3), F: SNORA52 (Small Nucleolar RNA, H/ACA Box 52), G: XKR5 (X Kell Blood Group Precursor-Related Family, Member 5), H: MED23 (Mediator Complex Subunit 23).

For evaluating the involvement of gender on DNA methylation, the β values of these representative eight genes were statistically evaluated. A significant difference in β values by gender was only identified by MED23 (P<0.05, Two-way ANOVA); however, the remaining seven genes showed no significant differences by gender. Furthermore, no significant differences in the interaction effect of disc degeneration (DD) groups and gender were also identified.

Identification of differentially methylated loci (DMLs) shared between human IVD degeneration and human knee osteoarthritis

When compared to data of 653 DMLs from human knee osteoarthritis cartilage[44], six common genes were identified (Table 3). Among 220 DMLs comprising a total of 187 individual genes found in the advanced stage of disc degeneration IVDs, 2.7% (6/220) DMLs, and 3.2% (6/187) individual genes were shared with those from knee OA cartilage previously reported by Alvarez-Garcia et al.[44].

Tab. 3.

Overlap of differentially methylated loci (DMLs) identified in human IVD degeneration and human knee osteoarthritis (OA).

<h2>Overlap of differentially methylated loci (DMLs) identified in human IVD degeneration and human knee osteoarthritis (OA).</h2>

Gene ontology analysis

The GO enrichment analysis of genes containing DMLs identified two significant GO terms for biological processes associated with cell adhesion; these were hemophilic cell adhesion through plasma membrane adhesion molecules (enrichment 11.2%, P = 5.86E-06) and cell-cell adhesion through plasma membrane adhesion molecules (enrichment 7.8%, P = 9.86E-05).

Discussion

This is the first study that compared DNA methylation profiles of the human NP between early and advanced stages of disc degeneration using the comprehensive methylation array, the Illumina Infinium MethylationEPIC array. We identified 220 differentially methylated loci that comprised a total of 187 individual genes, revealing that the early and advanced degenerated human NP tissues exhibit substantially different methylomes. Furthermore, the GO enrichment analysis identified two significant GO terms for biological processes associated with cell adhesion.

As written in the recent review on the biological aging of intervertebral disc[8], the authors described that disc degeneration could be theoretically distinguished from disc aging. Disc aging is considered to occur systemically in all spinal discs of all older individuals. On the other hand, no precise definition of "intervertebral disc degeneration" has been accepted for biomedical research and/or clinical practice. Adams et al.[7] reported that IVD degeneration is considered to be a structural failure with accelerated or advanced changes of the aging disc. Unfortunately, however, the specific biological differences between an aged disc and a degenerated disc have not been clearly defined because both share many similar biological, histological, and radiological changes[7]. Importantly, degenerative disc diseases also should be applied to be a degenerated disc that is also painful and/or associated with neurological symptoms[7].

In the clinical setting, human IVDs with a MRI finding of ‘no clear distinction between the AF and NP', which signifies the loss of signal of the NP on T2-weighted images, the generally accepted image findings of disc degeneration[45, 46], were assigned to Pfirrmann grade IV or V[32] (Fig 1). A previous study demonstrated that the loss of signal intensity in the NP area is significantly associated with the morphological features and biochemical contents of degenerative human IVDs[46]. Furthermore, the expression of catabolic factors, such as proinflammatory cytokines and matrix-degrading enzymes, was upregulated in the degenerated human IVD evaluated by MRI[4751]. We, therefore, defined an MRI classification of more than grade IV as ‘advanced stage of degeneration.'

Human IVD samples at the early stage of disc degeneration were obtained from anterior fusion surgeries of spinal trauma patients, except for one patient. On the other hand, those at the advanced stage of disc degeneration were obtained from spinal fusion surgeries for patients with degenerative lumbar diseases, such as lumbar spinal stenosis or degenerative spondylolisthesis. It should be kept in mind that the differences in DNA methylation profiles between these two groups, therefore, reflect not only changes in MRI findings, but also the underlying changes with/without lumbar degenerative diseases caused by progressive disc degeneration. Nevertheless, it would not be a practical issue in this study that the human IVD samples isolated from the patients with degenerative lumbar diseases can be regarded as the discs with advanced disc degeneration (AD).

Biochemical characteristics of IVD degeneration, especially those of NP tissues, have been characterized to represent the degradation of the extracellular matrix[9]. The biochemical changes of the major components of the human NP (type II collagen and the proteoglycan aggrecan), and also minor components, including collagen (types III, V, VI, IX-XII and XIV) and small proteoglycans (lumican, biglycan, decorin and fibromodulin), during disc degeneration have been well documented[9, 52]. However, the results of the current study showed no significant changes in DNA methylation profiles in these major and minor matrix components of human NP tissues between early and advanced disc degeneration.

Biologically, IVD cells, including NP cells, regulate the homeostasis of IVD tissues by maintaining a balance between anabolism and catabolism[9]. Therefore, an imbalance between anabolic and catabolic pathways is considered to be responsible for the onset and progression of IVD degeneration.

The progression of IVD degeneration is characterized by increased extracellular matrix degradation by locally produced matrix metalloproteinases (MMPs) and ADAMTSs (a disintegrin and metalloproteinase with thrombospondin motifs), which enzymatically degrade collagens and aggrecan. Importantly, the expression of those matrix-degrading enzymes can be stimulated by locally produced pro-inflammatory cytokines, such as interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α)[51, 5355]. However, the current study showed that MMPs, ADAMTSs, and proinflammatory cytokines were not differentially methylated in the advanced IVD degeneration stage compared to those in the early stage.

Activation of nuclear factor-κB (NF-κB), which plays a central role in inflammation through its ability to induce transcription of proinflammatory genes, including TNF-α, IL-1β, IL-6 and IL-8[56], has been shown to increase disc degeneration by upregulating the expression of matrix-degrading enzymes, such as MMPs and ADAMTSs[57]. Interestingly, we identified three hypermethylated genes in the advanced stage of disc degeneration (CARD14[58], EFHD2 and RTKN2[59]) that are involved in the regulation of the NF-κB pathway. Also, hypermethylated genes associated with the MAPK signaling pathway such as MAPKAPK5[60, 61] and PRKCZ[62] that have the potential to regulate multiple catabolic molecules were identified.

Importantly, the Wnt signaling pathway has also been reported to be associated with extracellular matrix metabolism by regulating pro-inflammatory stimuli. Our results showed that WNT5A, one of the Wnt proteins family, was differentially methylated in advanced stage degenerated IVD tissues. Wnt proteins are a major family of signal molecules that regulate cell biological and developmental processes[63]. Wnt proteins and the Wnt signaling pathway have also been implicated in the regulation of inflammatory processes in osteoarthritis and disc degeneration[6467]. Among Wnt proteins, Wnt-5a, a representative ligand that activates the β-catenin independent pathway in Wnt signaling, is involved in the pathogenesis of osteoarthritis (OA)[65, 66, 68].

Using immunohistological analysis, Li et al. reported that Wnt-5a was expressed in human NP tissues and that its expression was significantly elevated in degenerated human NP tissues[65]. Interestingly, recent studies showed that Wnt/β-catenin signaling pathway was activated by YAP1, a downstream nuclear effector of the Hippo signaling pathway[69, 70], which was also identified to be differentially methylated in the current study.

The results of the current study suggest the possibility that genes for catabolic molecules, including pro-inflammatory cytokines and matrix-degrading enzymes, may not be differentially methylated during disc degeneration in humans. However, DNA methylation may be differentially regulated in genes associated with signaling pathways, such as NF-κB, MAPK-ERK and Wnt signaling pathways, that are located upstream to the gene transcription of these catabolic molecules.

It is well known that the anabolic regulators of human IVD cells include polypeptide growth factors, such as IGF-1, transforming growth factor-β (TGF-β), and the bone morphogenetic proteins (BMPs)[71, 72].

SMADS, the main signal transducers for receptors of TGF-β[73], play important roles in stimulating cell proliferation and IVD cell matrix metabolism. We found that SMAD3 was differentially methylated in the IVD degeneration stage compared to that at the early stage. A previous study showed that TGF-β upregulated aggrecan and sulfated glycosaminoglycans (sGAG) synthesis [64]. sGAG synthesis was recently reported to be stimulated by the SAMD3 signaling pathway in the regulation of expression of β-1,3-glucuronosyltransferase 1 (GlcAT-1), a key enzyme that catalyzes glycosaminoglycan (GAG) synthesis[74, 75]. This suggests that SMAD3 may be implicated in IVD degeneration. Additionally, a differential methylation level of other genes that regulate TGF-β signaling, such as MECOM and ELAC2, was also identified in our study.

Interestingly, other important growth factor-related genes, such as IGFBP4 and FGFBP2, were found to be differentially methylated in the advanced IVD degeneration stage compared with the early stage. From these results, we can speculate that DNA methylation profiles may be differentially regulated, not only in catabolic factors, but also in anabolic factors, such as growth factors, that can regulate cell proliferation and extracellular matrix metabolism of the human NP.

Our results also showed that several enzymes that catalyze the biosynthesis of sulfated glycosaminoglycans (GAGs) of proteoglycans, including CHST1, EXTL3 and SLC26A2, were differentially methylated in the advanced stage of human disc degeneration compared to those in the early stage.

In our study, three genes associated with the regulation of Hedgehog (Hh) signaling were also found to be differentially methylated in the advanced IVD degeneration stage, including SUFU[7678], TTCIB[79] and IQCTH[80]. Hh signaling plays pivotal roles in regulating normal chondrocyte growth and differentiation. Lin et al. recently reported that a higher level of Hh signaling in chondrocytes is responsible for the severe osteoarthritis phenotype, suggesting that Hh signaling is associated with the severity of OA[81]. Furthermore, Sonic hedgehog (Shh), secreted by NP cells, is essential for cell proliferation in the growing disc and differentiation in the developmental stage of the mouse IVD[82]. Therefore, our results also suggest that changes in methylation profiles related to the hedgehog pathway may be responsible for the development of disc degeneration.

The GO enrichment analysis of differentially methylated genes further revealed significant GO terms for biological processes associated with cell adhesion; hemophilic cell adhesion through plasma membrane adhesion molecules and cell-cell adhesion through plasma membrane adhesion molecules. Cell-matrix interactions of NP cells, as well as chondrocytes, play crucial roles in regulating several functions, including cell survival and matrix metabolism, acting through anabolic and catabolic signaling pathways through integrin and other ECM receptors[8386]. The results of the current study suggest that differential methylation loci may not accumulate in ECM molecules and/or catabolic molecules themselves, but would rather accumulate in the molecules associated with cell-matrix and/or cell-cell adhesion that are related to the major signaling pathways relevant to the process of human disc degeneration.

Human IVDs and articular cartilage share remarkably similar anatomical composition, biochemical features and molecular processes of matrix degeneration. Genetically, these two matrix degenerative states also have common susceptibility alleles, such as single-nucleotide polymorphism rs143383 in the 5’ untranslated region of GDF5 and asporin D14 triplet repeat[87]. Therefore, we compared the DNA methylation profiles of human IVD degeneration (data from the current study) with those from a previous study of human knee OA[44]. The results of this analysis showed that 2.7% (6/220) of DMLs overlapped between these two diseases. The overlapping genes include YAP1[69, 70] and FGFRL1[8891], which have been reported to be associated with the pathogenesis of disc degeneration and OA. Although these two diseases share common pathological features, the DNA methylation profiles were very different. The authors speculate that, in addition to the genetic background, anatomical differences in the mobile joint structure of IVDs and knee joints would contribute to differences in DNA methylation profiles between these two diseases during the process of tissue degeneration.

There were some limitations to this study. First, IVD samples with early stage disc degeneration were difficult to obtain from spine surgeries. Most samples of MRI grades I to III were obtained from spinal trauma surgeries of relatively young patients. Because of the small number of samples, IVDs with Pfirrmann MRI grades I to III were all grouped as early stage degeneration. Differences in DNA methylation patterns among these three grades, which would be associated with the initiation of human disc degeneration, should be evaluated in a future study. Second, because the radiological, biochemical and histological features of degenerative changes in human NP tissues were well characterized[9], human NP tissues were isolated and processed for DNA methylation analysis in this study. On the other hand, AF tissues are also known to show degenerative changes, including irregularity of the lamella and collagen degeneration[9]. Therefore, it would also be of great importance to evaluate the DNA methylation profile of human AF tissues between early and advanced stages of disc degeneration in a future study. Third, recent epigenetic studies have shown that age and gender are significantly associated with the changes in DNA methylation profiles [9295]. Since differences in age and gender inequality exist between ED and AD group, there would be the possibility that these two factors may have potential to affect the DNA methylation profiles of human IVD degeneration in this study. Forth limitation of this study is that the expression of individual hypomethylated and hypermethylated genes was not examined in this study. It has been reported that gene transcription is also influenced by the gene features (CpG-dense promotors or gene body) where methylation occurs[13]. Therefore, the genome-wide gene expression analysis, such as RNA sequencing would be needed for further evaluating the function of DNA methylation in the process of human IVD degeneration in a future study.

Conclusion

We conducted, for the first time, a genome-wide DNA methylation profile comparative study and observed significant differences in DNA methylation profiles between early and advanced stages of human IVD degeneration.

The overview of the DNA methylation profile in the current study revealed that DMLs were identified in many genes associated with known molecules that have been reported to be relevant to IVD degeneration. Importantly, changes in DNA methylation profiles were also found in genes that regulate the major signaling pathways, such as NF-κB, MAPK, and Wnt signaling, that are well known to be responsible for the pathogenesis of human disc degeneration.

According to the GO analysis, DMLs tended to accumulate in molecules associated with cell adhesion, suggesting that diverse signaling pathways that regulate the cell-ECM or cell-cell interactions that orchestrate cell survival and matrix metabolism may be implicated in the process of human IVD degeneration.

Since the results of this study are still preliminary in a small number of samples, the evaluation of gene and protein expression in addition to a genome-wide DNA methylation profiles in an increasing number of samples would be needed to elucidate the pathological mechanism of human IVD degeneration in a future study.

Supporting information

S1 Table [xlsx]
Complete list of significantly differentially methylated loci (DMLs).

S1 Fig [tiff]
Differences in the β value of the four highest hypomethylated and hypermethylated loci among each grade of degeneration (Pfirrmann grades I-IV[]).


Zdroje

1. DALYs GBD, Collaborators H. Global, regional, and national disability-adjusted life-years (DALYs) for 315 diseases and injuries and healthy life expectancy (HALE), 1990–2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet. 2016;388(10053):1603–58. Epub 2016/10/14. doi: 10.1016/S0140-6736(16)31460-X 27733283; PubMed Central PMCID: PMC5388857.

2. Akeda K, Ohishi K, Masuda K, Bae WC, Takegami N, Yamada J, et al. Intradiscal Injection of Autologous Platelet-Rich Plasma Releasate to Treat Discogenic Low Back Pain: A Preliminary Clinical Trial. Asian Spine J. 2017;11(3):380–9. Epub 2017/07/04. doi: 10.4184/asj.2017.11.3.380 28670405; PubMed Central PMCID: PMC5481592.

3. DePalma MJ, Ketchum JM, Saullo T. What is the source of chronic low back pain and does age play a role? Pain Med. 2011;12(2):224–33. Epub 2011/01/27. doi: 10.1111/j.1526-4637.2010.01045.x 21266006.

4. Ohtori S, Inoue G, Miyagi M, Takahashi K. Pathomechanisms of discogenic low back pain in humans and animal models. Spine J. 2015;15(6):1347–55. Epub 2014/03/25. doi: 10.1016/j.spinee.2013.07.490 24657737.

5. Samartzis D, Karppinen J, Mok F, Fong DY, Luk KD, Cheung KM. A population-based study of juvenile disc degeneration and its association with overweight and obesity, low back pain, and diminished functional status. J Bone Joint Surg Am. 2011;93(7):662–70. Epub 2011/04/08. doi: 10.2106/JBJS.I.01568 21471420.

6. Suzuki H, Kanchiku T, Imajo Y, Yoshida Y, Nishida N, Taguchi T. Diagnosis and Characters of Non-Specific Low Back Pain in Japan: The Yamaguchi Low Back Pain Study. PLoS One. 2016;11(8):e0160454. Epub 2016/08/23. doi: 10.1371/journal.pone.0160454 27548658; PubMed Central PMCID: PMC4993356.

7. Adams MA, Roughley PJ. What is intervertebral disc degeneration, and what causes it? Spine (Phila Pa 1976). 2006;31(18):2151–61. doi: 10.1097/01.brs.0000231761.73859.2c 16915105.

8. Vo NV, Hartman RA, Patil PR, Risbud MV, Kletsas D, Iatridis JC, et al. Molecular mechanisms of biological aging in intervertebral discs. J Orthop Res. 2016;34(8):1289–306. doi: 10.1002/jor.23195 26890203; PubMed Central PMCID: PMC4988945.

9. Urban JP, Roberts S. Degeneration of the intervertebral disc. Arthritis Res Ther. 2003;5(3):120–30. Epub 2003/05/02. doi: 10.1186/ar629 12723977; PubMed Central PMCID: PMC165040.

10. Goldberg AD, Allis CD, Bernstein E. Epigenetics: a landscape takes shape. Cell. 2007;128(4):635–8. Epub 2007/02/27. doi: 10.1016/j.cell.2007.02.006 17320500.

11. Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003;33 Suppl:245–54. doi: 10.1038/ng1089 12610534.

12. Dor Y, Cedar H. Principles of DNA methylation and their implications for biology and medicine. Lancet. 2018;392(10149):777–86. Epub 2018/08/14. doi: 10.1016/S0140-6736(18)31268-6 30100054.

13. Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;13(7):484–92. doi: 10.1038/nrg3230 22641018.

14. Jin Z, Liu Y. DNA methylation in human diseases. Genes Dis. 2018;5(1):1–8. Epub 2018/09/28. doi: 10.1016/j.gendis.2018.01.002 30258928; PubMed Central PMCID: PMC6147084.

15. Sandoval J, Esteller M. Cancer epigenomics: beyond genomics. Curr Opin Genet Dev. 2012;22(1):50–5. doi: 10.1016/j.gde.2012.02.008 22402447.

16. Bui C, Barter MJ, Scott JL, Xu Y, Galler M, Reynard LN, et al. cAMP response element-binding (CREB) recruitment following a specific CpG demethylation leads to the elevated expression of the matrix metalloproteinase 13 in human articular chondrocytes and osteoarthritis. FASEB J. 2012;26(7):3000–11. doi: 10.1096/fj.12-206367 22505473.

17. Cheung KS, Hashimoto K, Yamada N, Roach HI. Expression of ADAMTS-4 by chondrocytes in the surface zone of human osteoarthritic cartilage is regulated by epigenetic DNA de-methylation. Rheumatol Int. 2009;29(5):525–34. doi: 10.1007/s00296-008-0744-z 18941754.

18. de Andres MC, Imagawa K, Hashimoto K, Gonzalez A, Roach HI, Goldring MB, et al. Loss of methylation in CpG sites in the NF-kappaB enhancer elements of inducible nitric oxide synthase is responsible for gene induction in human articular chondrocytes. Arthritis Rheum. 2013;65(3):732–42. doi: 10.1002/art.37806 23239081; PubMed Central PMCID: PMC3937961.

19. Hashimoto K, Oreffo RO, Gibson MB, Goldring MB, Roach HI. DNA demethylation at specific CpG sites in the IL1B promoter in response to inflammatory cytokines in human articular chondrocytes. Arthritis Rheum. 2009;60(11):3303–13. doi: 10.1002/art.24882 19877066; PubMed Central PMCID: PMC2788707.

20. Kim KI, Park YS, Im GI. Changes in the epigenetic status of the SOX-9 promoter in human osteoarthritic cartilage. J Bone Miner Res. 2013;28(5):1050–60. doi: 10.1002/jbmr.1843 23225119.

21. Reynard LN, Bui C, Syddall CM, Loughlin J. CpG methylation regulates allelic expression of GDF5 by modulating binding of SP1 and SP3 repressor proteins to the osteoarthritis susceptibility SNP rs143383. Hum Genet. 2014;133(8):1059–73. doi: 10.1007/s00439-014-1447-z 24861163; PubMed Central PMCID: PMC4099533.

22. Roach HI, Yamada N, Cheung KS, Tilley S, Clarke NM, Oreffo RO, et al. Association between the abnormal expression of matrix-degrading enzymes by human osteoarthritic chondrocytes and demethylation of specific CpG sites in the promoter regions. Arthritis Rheum. 2005;52(10):3110–24. doi: 10.1002/art.21300 16200590.

23. Scott JL, Gabrielides C, Davidson RK, Swingler TE, Clark IM, Wallis GA, et al. Superoxide dismutase downregulation in osteoarthritis progression and end-stage disease. Ann Rheum Dis. 2010;69(8):1502–10. doi: 10.1136/ard.2009.119966 20511611; PubMed Central PMCID: PMC3789136.

24. den Hollander W, Meulenbelt I. DNA Methylation in Osteoarthritis. Curr Genomics. 2015;16(6):419–26. doi: 10.2174/1389202916666150817212711 27019616; PubMed Central PMCID: PMC4765529.

25. den Hollander W, Ramos YF, Bos SD, Bomer N, van der Breggen R, Lakenberg N, et al. Knee and hip articular cartilage have distinct epigenomic landscapes: implications for future cartilage regeneration approaches. Ann Rheum Dis. 2014;73(12):2208–12. doi: 10.1136/annrheumdis-2014-205980 25261579.

26. Fernandez-Tajes J, Soto-Hermida A, Vazquez-Mosquera ME, Cortes-Pereira E, Mosquera A, Fernandez-Moreno M, et al. Genome-wide DNA methylation analysis of articular chondrocytes reveals a cluster of osteoarthritic patients. Ann Rheum Dis. 2014;73(4):668–77. doi: 10.1136/annrheumdis-2012-202783 23505229.

27. Jeffries MA, Donica M, Baker LW, Stevenson ME, Annan AC, Humphrey MB, et al. Genome-wide DNA methylation study identifies significant epigenomic changes in osteoarthritic cartilage. Arthritis Rheumatol. 2014;66(10):2804–15. doi: 10.1002/art.38762 24980887.

28. Moazedi-Fuerst FC, Hofner M, Gruber G, Weinhaeusel A, Stradner MH, Angerer H, et al. Epigenetic differences in human cartilage between mild and severe OA. J Orthop Res. 2014;32(12):1636–45. doi: 10.1002/jor.22722 25212754.

29. Rushton MD, Reynard LN, Barter MJ, Refaie R, Rankin KS, Young DA, et al. Characterization of the cartilage DNA methylome in knee and hip osteoarthritis. Arthritis Rheumatol. 2014;66(9):2450–60. doi: 10.1002/art.38713 24838673; PubMed Central PMCID: PMC4314681.

30. Zhang Y, Fukui N, Yahata M, Katsuragawa Y, Tashiro T, Ikegawa S, et al. Genome-wide DNA methylation profile implicates potential cartilage regeneration at the late stage of knee osteoarthritis. Osteoarthritis Cartilage. 2016;24(5):835–43. doi: 10.1016/j.joca.2015.12.013 26746145.

31. Zhao L, Wang Q, Zhang C, Huang C. Genome-wide DNA methylation analysis of articular chondrocytes identifies TRAF1, CTGF, and CX3CL1 genes as hypomethylated in osteoarthritis. Clin Rheumatol. 2017;36(10):2335–42. doi: 10.1007/s10067-017-3667-9 28470428.

32. Pfirrmann CW, Metzdorf A, Zanetti M, Hodler J, Boos N. Magnetic resonance classification of lumbar intervertebral disc degeneration. Spine (Phila Pa 1976). 2001;26(17):1873–8. Epub 2001/09/25. doi: 10.1097/00007632-200109010-00011 11568697.

33. Morris TJ, Butcher LM, Feber A, Teschendorff AE, Chakravarthy AR, Wojdacz TK, et al. ChAMP: 450k Chip Analysis Methylation Pipeline. Bioinformatics. 2014;30(3):428–30. Epub 2013/12/18. doi: 10.1093/bioinformatics/btt684 24336642; PubMed Central PMCID: PMC3904520.

34. Fortin JP, Triche TJ Jr., Hansen KD. Preprocessing, normalization and integration of the Illumina HumanMethylationEPIC array with minfi. Bioinformatics. 2017;33(4):558–60. Epub 2016/12/31. doi: 10.1093/bioinformatics/btw691 28035024; PubMed Central PMCID: PMC5408810.

35. Hansen K, Ayree M. minfi: Analyze Illumina’s 450k methylation arrays. R package version 1.8.3. 2011.

36. Maksimovic J, Gordon L, Oshlack A. SWAN: Subset-quantile within array normalization for illumina infinium HumanMethylation450 BeadChips. Genome Biol. 2012;13(6):R44. Epub 2012/06/19. doi: 10.1186/gb-2012-13-6-r44 22703947; PubMed Central PMCID: PMC3446316.

37. Zhou WD, Laird PW, Shen H. Comprehensive characterization, annotation and innovative use of Infinium DNA methylation BeadChip probes. Nucleic Acids Res. 2017;45(4). ARTN e22 doi: 10.1093/nar/gkw967 WOS:000396055400007. 27924034

38. Nordlund J, Backlin CL, Wahlberg P, Busche S, Berglund EC, Eloranta ML, et al. Genome-wide signatures of differential DNA methylation in pediatric acute lymphoblastic leukemia. Genome Biol. 2013;14(9):r105. Epub 2013/09/26. doi: 10.1186/gb-2013-14-9-r105 24063430; PubMed Central PMCID: PMC4014804.

39. Smyth GK. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 2004;3:Article3. Epub 2006/05/02. doi: 10.2202/1544-6115.1027 16646809.

40. Wettenhall JM, Smyth GK. limmaGUI: a graphical user interface for linear modeling of microarray data. Bioinformatics. 2004;20(18):3705–6. Epub 2004/08/07. doi: 10.1093/bioinformatics/bth449 15297296.

41. Geeleher P, Hartnett L, Egan LJ, Golden A, Ali RAR, Seoighe C. Gene-set analysis is severely biased when applied to genome-wide methylation data. Bioinformatics. 2013;29(15):1851–7. doi: 10.1093/bioinformatics/btt311 WOS:000322337000002. 23732277

42. Phipson B, Maksimovic J, Oshlack A. missMethyl: an R package for analyzing data from Illumina's HumanMethylation450 platform. Bioinformatics. 2016;32(2):286–8. doi: 10.1093/bioinformatics/btv560 WOS:000368360100018. 26424855

43. Young MD, Wakefield MJ, Smyth GK, Oshlack A. Gene ontology analysis for RNA-seq: accounting for selection bias. Genome Biology. 2010;11(2). ARTN R14 doi: 10.1186/gb-2010-11-2-r14 WOS:000276434300013. 20132535

44. Alvarez-Garcia O, Fisch KM, Wineinger NE, Akagi R, Saito M, Sasho T, et al. Increased DNA Methylation and Reduced Expression of Transcription Factors in Human Osteoarthritis Cartilage. Arthritis Rheumatol. 2016;68(8):1876–86. doi: 10.1002/art.39643 26881698; PubMed Central PMCID: PMC4963260.

45. Teraguchi M, Yoshimura N, Hashizume H, Muraki S, Yamada H, Minamide A, et al. Prevalence and distribution of intervertebral disc degeneration over the entire spine in a population-based cohort: the Wakayama Spine Study. Osteoarthritis Cartilage. 2014;22(1):104–10. Epub 2013/11/19. doi: 10.1016/j.joca.2013.10.019 24239943.

46. Benneker LM, Heini PF, Anderson SE, Alini M, Ito K. Correlation of radiographic and MRI parameters to morphological and biochemical assessment of intervertebral disc degeneration. Eur Spine J. 2005;14(1):27–35. Epub 2005/02/22. doi: 10.1007/s00586-004-0759-4 15723249; PubMed Central PMCID: PMC3476685.

47. Radek M, Pacholczyk-Sienicka B, Jankowski S, Albrecht L, Grodzka M, Depta A, et al. Assessing the correlation between the degree of disc degeneration on the Pfirrmann scale and the metabolites identified in HR-MAS NMR spectroscopy. Magn Reson Imaging. 2016;34(4):376–80. Epub 2015/12/29. doi: 10.1016/j.mri.2015.12.005 26708032.

48. Rodrigues LM, Oliveira LZ, Pinhal MA. Expression of heparanase isoforms in intervertebral discs classified according to Pfirrmann grading system for disc degeneration. Spine (Phila Pa 1976). 2013;38(13):1112–8. Epub 2013/02/02. doi: 10.1097/BRS.0b013e3182894cf4 23370684.

49. Iida R, Akeda K, Kasai Y, Masuda K, Morimoto R, Sakakibara T, et al. Expression of proteinase-activated receptor-2 in the intervertebral disc. Spine (Phila Pa 1976). 2009;34(5):470–8. doi: 10.1097/BRS.0b013e318195a67d 19247167.

50. Patel KP, Sandy JD, Akeda K, Miyamoto K, Chujo T, An HS, et al. Aggrecanases and aggrecanase-generated fragments in the human intervertebral disc at early and advanced stages of disc degeneration. Spine (Phila Pa 1976). 2007;32(23):2596–603. Epub 2007/11/06. doi: 10.1097/BRS.0b013e318158cb85 17978660.

51. 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(4):R77. Epub 2007/08/11. doi: 10.1186/ar2275 17688691; PubMed Central PMCID: PMC2206382.

52. Zhao CQ, Wang LM, Jiang LS, Dai LY. The cell biology of intervertebral disc aging and degeneration. Ageing Res Rev. 2007;6(3):247–61. Epub 2007/09/18. doi: 10.1016/j.arr.2007.08.001 17870673.

53. Le Maitre CL, Pockert A, Buttle DJ, Freemont AJ, Hoyland JA. Matrix synthesis and degradation in human intervertebral disc degeneration. Biochem Soc Trans. 2007;35(Pt 4):652–5. doi: 10.1042/BST0350652 17635113.

54. Nakki A, Battie MC, Kaprio J. Genetics of disc-related disorders: current findings and lessons from other complex diseases. Eur Spine J. 2014;23 Suppl 3:S354–63. Epub 2013/07/11. doi: 10.1007/s00586-013-2878-2 23838702.

55. Wang SZ, Rui YF, Lu J, Wang C. Cell and molecular biology of intervertebral disc degeneration: current understanding and implications for potential therapeutic strategies. Cell Prolif. 2014;47(5):381–90. doi: 10.1111/cpr.12121 25112472.

56. Tak PP, Firestein GS. NF-kappaB: a key role in inflammatory diseases. J Clin Invest. 2001;107(1):7–11. Epub 2001/01/03. doi: 10.1172/JCI11830 11134171; PubMed Central PMCID: PMC198552.

57. Nerlich AG, Bachmeier BE, Schleicher E, Rohrbach H, Paesold G, Boos N. Immunomorphological analysis of RAGE receptor expression and NF-kappaB activation in tissue samples from normal and degenerated intervertebral discs of various ages. Ann N Y Acad Sci. 2007;1096:239–48. Epub 2007/04/05. doi: 10.1196/annals.1397.090 17405935.

58. Zotti T, Polvere I, Voccola S, Vito P, Stilo R. CARD14/CARMA2 Signaling and its Role in Inflammatory Skin Disorders. Front Immunol. 2018;9:2167. Epub 2018/10/16. doi: 10.3389/fimmu.2018.02167 30319628; PubMed Central PMCID: PMC6168666.

59. Myouzen K, Kochi Y, Okada Y, Terao C, Suzuki A, Ikari K, et al. Functional variants in NFKBIE and RTKN2 involved in activation of the NF-kappaB pathway are associated with rheumatoid arthritis in Japanese. PLoS Genet. 2012;8(9):e1002949. Epub 2012/10/03. doi: 10.1371/journal.pgen.1002949 23028356; PubMed Central PMCID: PMC3441678.

60. Ni H, Wang XS, Diener K, Yao Z. MAPKAPK5, a novel mitogen-activated protein kinase (MAPK)-activated protein kinase, is a substrate of the extracellular-regulated kinase (ERK) and p38 kinase. Biochem Biophys Res Commun. 1998;243(2):492–6. Epub 1998/03/03. doi: 10.1006/bbrc.1998.8135 9480836.

61. Westhovens R, Keyser FD, Rekalov D, Nasonov EL, Beetens J, Van der Aa A, et al. Oral administration of GLPG0259, an inhibitor of MAPKAPK5, a new target for the treatment of rheumatoid arthritis: a phase II, randomised, double-blind, placebo-controlled, multicentre trial. Ann Rheum Dis. 2013;72(5):741–4. Epub 2012/11/20. doi: 10.1136/annrheumdis-2012-202221 23161899.

62. Monick MM, Carter AB, Flaherty DM, Peterson MW, Hunninghake GW. Protein kinase C zeta plays a central role in activation of the p42/44 mitogen-activated protein kinase by endotoxin in alveolar macrophages. J Immunol. 2000;165(8):4632–9. Epub 2000/10/18. doi: 10.4049/jimmunol.165.8.4632 11035106.

63. Komiya Y, Habas R. Wnt signal transduction pathways. Organogenesis. 2008;4(2):68–75. Epub 2009/03/13. doi: 10.4161/org.4.2.5851 19279717; PubMed Central PMCID: PMC2634250.

64. De Santis M, Di Matteo B, Chisari E, Cincinelli G, Angele P, Lattermann C, et al. The Role of Wnt Pathway in the Pathogenesis of OA and Its Potential Therapeutic Implications in the Field of Regenerative Medicine. Biomed Res Int. 2018;2018:7402947. Epub 2018/11/10. doi: 10.1155/2018/7402947 30410938; PubMed Central PMCID: PMC6205317.

65. Li Z, Zhang K, Li X, Pan H, Li S, Chen F, et al. Wnt5a suppresses inflammation-driven intervertebral disc degeneration via a TNF-alpha/NF-kappaB-Wnt5a negative-feedback loop. Osteoarthritis Cartilage. 2018;26(7):966–77. Epub 2018/04/16. doi: 10.1016/j.joca.2018.04.002 29656141.

66. Ryu JH, Chun JS. Opposing roles of WNT-5A and WNT-11 in interleukin-1beta regulation of type II collagen expression in articular chondrocytes. J Biol Chem. 2006;281(31):22039–47. Epub 2006/06/07. doi: 10.1074/jbc.M601804200 16754689.

67. Smolders LA, Meij BP, Onis D, Riemers FM, Bergknut N, Wubbolts R, et al. Gene expression profiling of early intervertebral disc degeneration reveals a down-regulation of canonical Wnt signaling and caveolin-1 expression: implications for development of regenerative strategies. Arthritis Res Ther. 2013;15(1):R23. Epub 2013/01/31. doi: 10.1186/ar4157 23360510; PubMed Central PMCID: PMC3672710.

68. Ge XP, Gan YH, Zhang CG, Zhou CY, Ma KT, Meng JH, et al. Requirement of the NF-kappaB pathway for induction of Wnt-5A by interleukin-1beta in condylar chondrocytes of the temporomandibular joint: functional crosstalk between the Wnt-5A and NF-kappaB signaling pathways. Osteoarthritis Cartilage. 2011;19(1):111–7. Epub 2010/11/03. doi: 10.1016/j.joca.2010.10.016 21035559.

69. Chen J, Mei Z, Huang B, Zhang X, Liu J, Shan Z, et al. IL-6/YAP1/beta-catenin signaling is involved in intervertebral disc degeneration. J Cell Physiol. 2018. Epub 2018/12/05. doi: 10.1002/jcp.27065 30511395.

70. Yang B, Sun H, Song F, Yu M, Wu Y, Wang J. YAP1 negatively regulates chondrocyte differentiation partly by activating the beta-catenin signaling pathway. Int J Biochem Cell Biol. 2017;87:104–13. Epub 2017/04/26. doi: 10.1016/j.biocel.2017.04.007 28438716.

71. Masuda K, An HS. Growth factors and the intervertebral disc. Spine J. 2004;4(6 Suppl):330S–40S. Epub 2004/11/16. doi: 10.1016/j.spinee.2004.07.028 15541686.

72. Masuda K, Imai Y, Okuma M, Muehleman C, Nakagawa K, Akeda K, et al. Osteogenic protein-1 injection into a degenerated disc induces the restoration of disc height and structural changes in the rabbit anular puncture model. Spine (Phila Pa 1976). 2006;31(7):742–54. doi: 10.1097/01.brs.0000206358.66412.7b 16582847.

73. Li TF, O'Keefe RJ, Chen D. TGF-beta signaling in chondrocytes. Front Biosci. 2005;10:681–8. Epub 2004/12/01. doi: 10.2741/1563 15569609; PubMed Central PMCID: PMC2647990.

74. Hu B, Xu C, Cao P, Tian Y, Zhang Y, Shi C, et al. TGF-beta Stimulates Expression of Chondroitin Polymerizing Factor in Nucleus Pulposus Cells Through the Smad3, RhoA/ROCK1, and MAPK Signaling Pathways. J Cell Biochem. 2018;119(1):566–79. Epub 2017/06/14. doi: 10.1002/jcb.26215 28608941.

75. Wu Q, Wang J, Skubutyte R, Kepler CK, Huang Z, Anderson DG, et al. Smad3 controls beta-1,3-glucuronosyltransferase 1 expression in rat nucleus pulposus cells: implications of dysregulated expression in disc disease. Arthritis Rheum. 2012;64(10):3324–33. Epub 2012/06/08. doi: 10.1002/art.34570 22674034; PubMed Central PMCID: PMC3601452.

76. Cheng SY, Yue S. Role and regulation of human tumor suppressor SUFU in Hedgehog signaling. Adv Cancer Res. 2008;101:29–43. Epub 2008/12/06. doi: 10.1016/S0065-230X(08)00402-8 19055941.

77. Tukachinsky H, Lopez LV, Salic A. A mechanism for vertebrate Hedgehog signaling: recruitment to cilia and dissociation of SuFu-Gli protein complexes. J Cell Biol. 2010;191(2):415–28. Epub 2010/10/20. doi: 10.1083/jcb.201004108 20956384; PubMed Central PMCID: PMC2958481.

78. Zhou F, Huang D, Li Y, Hu G, Rao H, Lu Q, et al. Nek2A/SuFu feedback loop regulates Gli-mediated Hedgehog signaling pathway. Int J Oncol. 2017;50(2):373–80. Epub 2016/12/31. doi: 10.3892/ijo.2016.3819 28035348; PubMed Central PMCID: PMC5238777.

79. Stottmann RW, Tran PV, Turbe-Doan A, Beier DR. Ttc21b is required to restrict sonic hedgehog activity in the developing mouse forebrain. Dev Biol. 2009;335(1):166–78. Epub 2009/09/08. doi: 10.1016/j.ydbio.2009.08.023 19732765; PubMed Central PMCID: PMC2778284.

80. Pusapati GV, Hughes CE, Dorn KV, Zhang D, Sugianto P, Aravind L, et al. EFCAB7 and IQCE regulate hedgehog signaling by tethering the EVC-EVC2 complex to the base of primary cilia. Dev Cell. 2014;28(5):483–96. Epub 2014/03/04. doi: 10.1016/j.devcel.2014.01.021 24582806; PubMed Central PMCID: PMC4027042.

81. Lin AC, Seeto BL, Bartoszko JM, Khoury MA, Whetstone H, Ho L, et al. Modulating hedgehog signaling can attenuate the severity of osteoarthritis. Nat Med. 2009;15(12):1421–5. Epub 2009/11/17. doi: 10.1038/nm.2055 19915594.

82. Dahia CL, Mahoney E, Wylie C. Shh signaling from the nucleus pulposus is required for the postnatal growth and differentiation of the mouse intervertebral disc. PLoS One. 2012;7(4):e35944. Epub 2012/05/05. doi: 10.1371/journal.pone.0035944 22558278; PubMed Central PMCID: PMC3338762.

83. Aota Y, An HS, Homandberg G, Thonar EJ, Andersson GB, Pichika R, et al. Differential effects of fibronectin fragment on proteoglycan metabolism by intervertebral disc cells: a comparison with articular chondrocytes. Spine (Phila Pa 1976). 2005;30(7):722–8. Epub 2005/04/02. doi: 10.1097/01.brs.0000157417.59933.db 15803072.

84. Gao Y, Liu S, Huang J, Guo W, Chen J, Zhang L, et al. The ECM-cell interaction of cartilage extracellular matrix on chondrocytes. Biomed Res Int. 2014;2014:648459. Epub 2014/06/25. doi: 10.1155/2014/648459 24959581; PubMed Central PMCID: PMC4052144.

85. Gilchrist CL, Chen J, Richardson WJ, Loeser RF, Setton LA. Functional integrin subunits regulating cell-matrix interactions in the intervertebral disc. J Orthop Res. 2007;25(6):829–40. Epub 2007/02/24. doi: 10.1002/jor.20343 17318895.

86. Gilchrist CL, Francisco AT, Plopper GE, Chen J, Setton LA. Nucleus pulposus cell-matrix interactions with laminins. Eur Cell Mater. 2011;21:523–32. Epub 2011/06/29. 21710443; PubMed Central PMCID: PMC3332080.

87. Loughlin J. Knee osteoarthritis, lumbar-disc degeneration and developmental dysplasia of the hip—an emerging genetic overlap. Arthritis Res Ther. 2011;13(2):108. Epub 2011/05/06. doi: 10.1186/ar3291 21542882; PubMed Central PMCID: PMC3132037.

88. Nagano T, Yonenobu K, Miyamoto S, Tohyama M, Ono K. Distribution of the basic fibroblast growth factor and its receptor gene expression in normal and degenerated rat intervertebral discs. Spine (Phila Pa 1976). 1995;20(18):1972–8. Epub 1995/09/15. doi: 10.1097/00007632-199509150-00002 8578370.

89. Weng T, Yi L, Huang J, Luo F, Wen X, Du X, et al. Genetic inhibition of fibroblast growth factor receptor 1 in knee cartilage attenuates the degeneration of articular cartilage in adult mice. Arthritis Rheum. 2012;64(12):3982–92. Epub 2012/07/27. doi: 10.1002/art.34645 22833219; PubMed Central PMCID: PMC3690192.

90. Xu W, Xie Y, Wang Q, Wang X, Luo F, Zhou S, et al. A novel fibroblast growth factor receptor 1 inhibitor protects against cartilage degradation in a murine model of osteoarthritis. Sci Rep. 2016;6:24042. Epub 2016/04/05. doi: 10.1038/srep24042 27041213; PubMed Central PMCID: PMC4819196.

91. Yan D, Chen D, Cool SM, van Wijnen AJ, Mikecz K, Murphy G, et al. Fibroblast growth factor receptor 1 is principally responsible for fibroblast growth factor 2-induced catabolic activities in human articular chondrocytes. Arthritis Res Ther. 2011;13(4):R130. Epub 2011/08/13. doi: 10.1186/ar3441 21835001; PubMed Central PMCID: PMC3239372.

92. Ciccarone F, Tagliatesta S, Caiafa P, Zampieri M. DNA methylation dynamics in aging: how far are we from understanding the mechanisms? Mech Ageing Dev. 2018;174:3–17. Epub 2017/12/23. doi: 10.1016/j.mad.2017.12.002 29268958.

93. Gopalan S, Carja O, Fagny M, Patin E, Myrick JW, McEwen LM, et al. Trends in DNA Methylation with Age Replicate Across Diverse Human Populations. Genetics. 2017;206(3):1659–74. Epub 2017/05/24. doi: 10.1534/genetics.116.195594 28533441; PubMed Central PMCID: PMC5500158.

94. McCarthy MM, Auger AP, Bale TL, De Vries GJ, Dunn GA, Forger NG, et al. The epigenetics of sex differences in the brain. J Neurosci. 2009;29(41):12815–23. Epub 2009/10/16. doi: 10.1523/JNEUROSCI.3331-09.2009 19828794; PubMed Central PMCID: PMC2788155.

95. Xia Y, Dai R, Wang K, Jiao C, Zhang C, Xu Y, et al. Sex-differential DNA methylation and associated regulation networks in human brain implicated in the sex-biased risks of psychiatric disorders. Mol Psychiatry. 2019. Epub 2019/04/13. doi: 10.1038/s41380-019-0416-2 30976086.


Článek vyšel v časopise

PLOS One


2019 Číslo 9
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Důležitost adherence při depresivním onemocnění
nový kurz
Autoři: MUDr. Eliška Bartečková, Ph.D.

Svět praktické medicíny 1/2024 (znalostní test z časopisu)

Koncepce osteologické péče pro gynekology a praktické lékaře
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#