Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Mol Med ; 30(1): 57, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698308

ABSTRACT

BACKGROUND: Ossification of the posterior longitudinal ligament (OPLL), an emerging heterotopic ossification disease, causes spinal cord compression, resulting in motor and sensory dysfunction. The etiology of OPLL remains unclear but may involve integrin αVß3 regulating the process of osteogenesis and angiogenesis. In this study, we focused on the role of integrin αVß3 in OPLL and explored the underlying mechanism by which the c(RGDyk) peptide acts as a potent and selective integrin αVß3 inhibitor to inhibit osteogenesis and angiogenesis in OPLL. METHODS: OPLL or control ligament samples were collected in surgery. For OPLL samples, RNA-sequencing results revealed activation of the integrin family, particularly integrin αVß3. Integrin αVß3 expression was detected by qPCR, Western blotting, and immunohistochemical analysis. Fluorescence microscopy was used to observe the targeted inhibition of integrin αVß3 by the c(RGDyk) peptide on ligaments fibroblasts (LFs) derived from patients with OPLL and endothelial cells (ECs). The effect of c(RGDyk) peptide on the ossification of pathogenic LFs was detected using qPCR, Western blotting. Alkaline phosphatase staining or alizarin red staining were used to test the osteogenic capability. The effect of the c(RGDyk) peptide on angiogenesis was determined by EC migration and tube formation assays. The effects of the c(RGDyk) peptide on heterotopic bone formation were evaluated by micro-CT, histological, immunohistochemical, and immunofluorescence analysis in vivo. RESULTS: The results indicated that after being treated with c(RGDyk), the osteogenic differentiation of LFs was significantly decreased. Moreover, the c(RGDyk) peptide inhibited the migration of ECs and thus prevented the nutritional support required for osteogenesis. Furthermore, the c(RGDyk) peptide inhibited ectopic bone formation in mice. Mechanistic analysis revealed that c(RGDyk) peptide could inhibit osteogenesis and angiogenesis in OPLL by targeting integrin αVß3 and regulating the FAK/ERK pathway. CONCLUSIONS: Therefore, the integrin αVß3 appears to be an emerging therapeutic target for OPLL, and the c(RGDyk) peptide has dual inhibitory effects that may be valuable for the new therapeutic strategy of OPLL.


Subject(s)
Integrin alphaVbeta3 , Ossification of Posterior Longitudinal Ligament , Osteogenesis , Integrin alphaVbeta3/metabolism , Integrin alphaVbeta3/antagonists & inhibitors , Humans , Osteogenesis/drug effects , Animals , Mice , Ossification of Posterior Longitudinal Ligament/metabolism , Ossification of Posterior Longitudinal Ligament/drug therapy , Male , Female , Middle Aged , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Neovascularization, Physiologic/drug effects , Cell Movement/drug effects , Disease Models, Animal , Oligopeptides/pharmacology , Oligopeptides/chemistry , Angiogenesis
2.
Am J Hum Genet ; 110(4): 638-647, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36990086

ABSTRACT

Ossification of the posterior longitudinal ligament of the spine (OPLL) is a common intractable disease that causes spinal stenosis and myelopathy. We have previously conducted genome-wide association studies for OPLL and identified 14 significant loci, but their biological implications remain mostly unclear. Here, we examined the 12p11.22 locus and identified a variant in the 5' UTR of a novel isoform of CCDC91 that was associated with OPLL. Using machine learning prediction models, we determined that higher expression of the novel CCDC91 isoform was associated with the G allele of rs35098487. The risk allele of rs35098487 showed higher affinity in the binding of nuclear proteins and transcription activity. Knockdown and overexpression of the CCDC91 isoform in mesenchymal stem cells and MG-63 cells showed paralleled expression of osteogenic genes, including RUNX2, the master transcription factor of osteogenic differentiation. The CCDC91 isoform directly interacted with MIR890, which bound to RUNX2 and decreased RUNX2 expression. Our findings suggest that the CCDC91 isoform acts as a competitive endogenous RNA by sponging MIR890 to increase RUNX2 expression.


Subject(s)
Ossification of Posterior Longitudinal Ligament , Osteogenesis , Humans , Osteogenesis/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Genome-Wide Association Study , Ossification of Posterior Longitudinal Ligament/genetics , Ossification of Posterior Longitudinal Ligament/metabolism , RNA, Untranslated
3.
Eur Cell Mater ; 45: 31-45, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36749152

ABSTRACT

The pathogenesis of posterior longitudinal ligament ossification (OPLL) remains inadequately understood. Mechanical stimulation is one of the important pathogenic factors in OPLL. As one of the mechanical stimulation transduction signals, the yes-associated protein (YAP) interacts with the Wnt/ß-catenin signalling pathway, which plays an important role in osteogenic differentiation. This study aimed to demonstrate the role of YAP-Wnt/ß-catenin axis in cell differentiation induced by mechanical stress. Primary cells extracted from posterior longitudinal ligament tissues from OPLL or non-OPLL patients were subjected to sinusoidal uniaxial cyclic stretch (5 %, 0.5 Hz, 3 d). The expression of runt-related transcription factor 2, collagen I, osterix, osteocalcin and alkaline phosphatase were compared between the static and the experimental groups. In addition, the cytoskeleton was detected using phalloidin staining while YAP phosphorylation states and nuclear location were identified using immunofluorescence. The results showed that mechanical stretching loading increased the expression of osteogenic genes and proteins in the OPLL group, while it had no significant effect on the control group. When OPLL cells were stretched, YAP exhibited an obvious nuclear translocation and the Wnt/ß-catenin pathway was activated. Knocking down YAP or ß-catenin could weaken the impact upon osteogenic differentiation induced by mechanical stimulation. YAP-mediated mechanical stimulation promoted osteogenic differentiation of OPLL cells through Wnt/ß-catenin pathway and this progress was independent of the Hippo pathway.


Subject(s)
Ossification of Posterior Longitudinal Ligament , Osteogenesis , Humans , beta Catenin/metabolism , Cell Differentiation , Cells, Cultured , Ossification of Posterior Longitudinal Ligament/metabolism , Ossification of Posterior Longitudinal Ligament/pathology , Wnt Signaling Pathway
4.
Cell Tissue Res ; 391(1): 145-157, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36305971

ABSTRACT

Interleukin-6 (IL-6) has been reported to induce osteogenic differentiation of mesenchymal stem cells for increasing bone regeneration, while the role of IL-6 in osteogenic differentiation during ossification of the posterior longitudinal ligament (OPLL) remains to be determined. The current study aims to explore the downstream mechanism of IL-6 in cyclic tensile strain (CTS)-stimulated OPLL, which involves bioinformatically identified microRNA-135b (miR-135b). Initially, we clinically collected posterior longitudinal ligament (PLL) and ossified PLL tissues, from which ossified PLL cells were isolated, respectively. The obtained data revealed a greater osteogenic property of ossified PLL than non-ossified PLL cells. The effect of regulatory axis comprising IL-6, Stat3, miR-135b, and BMPER on osteogenic differentiation of CTS-stimulated ossified PLL cells was examined with gain- and loss-of-function experiments. BMPER was confirmed as a target gene to miR-135b. Knockdown of BMPER or overexpression of miR-135b inhibited the osteogenic differentiation of CTS-induced ossification in PLL cells. Besides, IL-6 promoted the post-transcriptional process to mature miR-135b via Stat3 phosphorylation. In conclusion, IL-6 inhibited CTS-induced osteogenic differentiation by inducing miR-135b-mediated inhibition of BMPER through Stat3 activation.


Subject(s)
Interleukin-6 , MicroRNAs , Ossification of Posterior Longitudinal Ligament , STAT3 Transcription Factor , Humans , Carrier Proteins , Cell Differentiation/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Longitudinal Ligaments , MicroRNAs/genetics , Ossification of Posterior Longitudinal Ligament/genetics , Ossification of Posterior Longitudinal Ligament/metabolism , Osteogenesis/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
5.
Oxid Med Cell Longev ; 2022: 1604932, 2022.
Article in English | MEDLINE | ID: mdl-35391932

ABSTRACT

Connexin 43- (Cx43-) mediated nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB) signaling has been found involved in the ossification of the posterior longitudinal ligament (OPLL). However, the underlying mechanism how OPLL is regulated has not been elucidated. In the present study, primary ligament fibroblast were isolated; immunoprecipitation (IP) and liquid chromatography-mass spectrometry (LC-MS) assays were applied to identify potential binding proteins of Cx43. Protein interaction was then confirmed by co-IP assay. Alkaline phosphatase (ALP) activity and alizarin red staining were used to evaluate ossification. Luciferase reporter assay and chromatin immunoprecipitation (ChIP) assay were employed to assess the binding between NF-κB p65 and target gene. Lipoxygenase inhibitor (5,8,11-eicosatriynoic acid, EPA) was applied to induce endoplasmic reticulum (ER) stress, and 4-phenylbutyrate (4-PBA) was used as an ER-stress inhibitor. Expression of USP9X, Cx43, and nuclei p65 in ligaments from patients and controls was detected by Western blotting. The results showed that ubiquitin-specific protease 9 X-chromosome (USP9X), a deubiquitylating enzyme, was a candidate of Cx43 binding proteins, and USP9X inhibited Cx43 ubiquitination. In vitro experiments showed that USP9X promoted ossification of primary ligament fibroblasts and nuclear translocation of NF-κB p65 by regulating Cx43 expression. Moreover, NF-κB can bind to the USP9X promoter to promote its transcription. When ER stress was inhibited by 4-PBA, USP9X levels, NF-κB nuclei translocation, and ALP activity were decreased. Reverse results were obtained when ER stress was induced by EPA. PDTC, an NF-κB inhibitor, could abolish the effects of EPA. Furthermore, USP9X, Cx43, and nuclei p65 were significantly upregulated in ligaments from OPLL patients than non-OPLL controls. USP9X was positively correlated with CX43 and nuclei p65 in OPLL samples. Overall, the findings suggest that the ER stress-NFκB-USP9X-Cx43 signaling pathway plays an important role in OPLL progression.


Subject(s)
Connexin 43 , Ossification of Posterior Longitudinal Ligament , Transcription Factor RelA , Ubiquitin Thiolesterase , Cells, Cultured , Cervical Vertebrae/metabolism , Connexin 43/genetics , Connexin 43/metabolism , Humans , Longitudinal Ligaments/metabolism , NF-kappa B/metabolism , Ossification of Posterior Longitudinal Ligament/genetics , Ossification of Posterior Longitudinal Ligament/metabolism , Osteogenesis/genetics , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Specific Proteases/metabolism
6.
Aging (Albany NY) ; 13(16): 20192-20217, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34483137

ABSTRACT

Ossification of the posterior longitudinal ligament (OPLL) is a disorder with multiple pathogenic mechanisms and leads to different degrees of neurological symptoms. Recent studies have revealed that non-coding RNA (ncRNA), including long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), could influence the development of OPLL. Nevertheless, the molecular mechanisms linking circular RNAs (circRNAs) and the progression of OPLL is still unknown. The current research explored the expression profiles of OPLL-related circRNAs by microarray analysis, and applied qRT-PCR to validate the results. Subsequently, we confirmed the upregulation of hsa_circ_0007292 in OPLL cells by qRT-PCR and validated the circular characteristic of hsa_circ_0007292 by Sanger sequencing. Fluorescence in situ hybridization (FISH) unveiled that hsa_circ_0007292 was predominantly located in the cytoplasm. Functionally, gain-of-function and loss-of-function experiments showed that hsa_circ_0007292 promoted the osteogenic differentiation of OPLL cells. Mechanistically, the interaction of hsa_circ_0007292 and miR-508-3p was predicted and validated by bioinformatics analysis, dual-luciferase reporter assays, and Ago2 RNA immunoprecipitation (RIP). Similarly, we validated the correlation between miR-508-3p and SATB2. Furthermore, rescue experiments were performed to prove that hsa_circ_0007292 acted as a sponge for miR-508-3p, and SATB2 was revealed to be the target gene of miR-508-3p. In conclusion, our research shows that hsa_circ_0007292 regulates OPLL progression by the miR-508-3p/SATB2 pathway. Our results indicate that hsa_circ_0007292 can be used as a promising therapeutic target for patients with OPLL.


Subject(s)
Matrix Attachment Region Binding Proteins/genetics , MicroRNAs/metabolism , Ossification of Posterior Longitudinal Ligament/metabolism , Osteogenesis , RNA, Circular/metabolism , Transcription Factors/genetics , Cell Line, Tumor , Cell Proliferation , Humans , Longitudinal Ligaments/cytology , Longitudinal Ligaments/metabolism , Matrix Attachment Region Binding Proteins/metabolism , MicroRNAs/genetics , Ossification of Posterior Longitudinal Ligament/genetics , Ossification of Posterior Longitudinal Ligament/physiopathology , RNA, Circular/genetics , Transcription Factors/metabolism , Up-Regulation
7.
Life Sci ; 279: 119481, 2021 Aug 15.
Article in English | MEDLINE | ID: mdl-33857573

ABSTRACT

AIMS: Muscle and adipose tissue-derived mesenchymal stem cells presented high osteogenic potentials, which modulate osteoblast function through releasing extracellular vesicles (EVs) containing miRNAs. Herein, this study evaluated the function of bone marrow mesenchymal stem cell-derived extracellular vesicles (BMSC-EVs) delivering miR-497-5p in ossification of the posterior longitudinal ligament (OPLL). MAIN METHODS: The expression level of miR-497-5p was validated in ossified posterior longitudinal ligament (PLL) tissues and BMSC-EVs. The uptake of BMSC-EVs by ligament fibroblasts was observed by immunofluorescence. miR-497-5p was overexpressed or downregulated to assess its role in osteogenic differentiation of ligament fibroblasts. Further, an OPLL rat model was established to substantiate the effect of BMSC-EVs enriched with miR-497-5p on OPLL. KEY FINDINGS: Ossified PLL tissues presented with high miR-497-5p expression. PLL fibroblasts were identified to endocytose BMSC-EVs. BMSC-EVs could upregulate miR-497-5p and shuttle it to ligament fibroblasts to accelerate the osteogenic differentiation. miR-497-5p targeted and inversely regulated RSPO2. Then, RSPO2 overexpression activated Wnt/ß-catenin pathway and repressed the osteogenic differentiation of ligament fibroblasts. In vivo experiments further showed that miR-497-5p-containing BMSC-EVs enhanced OPLL through diminishing RSPO2 and inactivating Wnt/ß-catenin pathway. SIGNIFICANCE: BMSC-EVs could deliver miR-497-5p to ligament fibroblasts and modulate RSPO2-mediated Wnt/ß-catenin pathway, thereby accelerating OPLL.


Subject(s)
Cell Differentiation , Extracellular Vesicles/pathology , Gene Expression Regulation , Mesenchymal Stem Cells/pathology , MicroRNAs/genetics , Ossification of Posterior Longitudinal Ligament/pathology , Thrombospondins/antagonists & inhibitors , Animals , Extracellular Vesicles/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Ossification of Posterior Longitudinal Ligament/genetics , Ossification of Posterior Longitudinal Ligament/metabolism , Rats , Rats, Wistar , Thrombospondins/genetics , Thrombospondins/metabolism
8.
Spine (Phila Pa 1976) ; 45(22): E1460-E1468, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32756283

ABSTRACT

STUDY DESIGN: Immunohistochemical and real-time reverse transcription-polymerase chain reaction (RT-PCR) analysis. OBJECTIVE: The aim of this study was to analyze the expression of five susceptibility genes (RSPO2, HAO1, CCDC91, RHPH9, and STK38L) for human ossification of the posterior longitudinal ligaments (OPLL) identified in a genome-wide association study. SUMMARY OF BACKGROUND DATA: Detailed expression and functional studies for the five susceptibility genes are needed to aid in clarification of the etiology and pathogenesis of OPLL. METHODS: Immunostaining, cell culture, and real-time RT-PCR were performed on ossified ligament samples collected during anterior cervical decompression for symptomatic OPLL (n = 39 patients) and on control non-OPLL samples (n = 8 patients). Immunohistochemical analysis in spinal hyperostotic mice (ttw/ttw) (n = 25) was also performed. The sample sections were stained for RSPO2, HAO1, CCDC91, RHPH9, STK38L, Runx2, Sox9, and CD90. The mRNA expression levels of the five susceptibility genes were also analyzed in cultured human OPLL and non-OPLL cells subjected to cyclic tensile strain. RESULTS: Immunoreactivity for RSPO2 and Sox9 was evident in proliferating chondrocytes in human OPLL tissues and ttw/ttw mice. Application of cyclic tensile strain to cultured human OPLL cells resulted in increases in mRNA levels for RSPO2, HAO1, and CCDC91. However, individual differences in expression in human OPLL-related samples were seen. HAO1-positive cells were detected only in 3- to 6-week-old ttw/ttw mice that did not simultaneously express RSPO2-positive samples. CONCLUSION: Among the five susceptibility genes, RSPO2, HAO1, and CCDC91 might be contributory factors in progression of OPLL. RSPO2 may be involved in endochondral ossification, especially in mixed or continuous type OPLL, HAO1 may be an initiation factor for OPLL that is rarely seen in mature human OPLL samples, and CCDC91 may be associated with progression of ossification caused by mechanical stress. These findings provide important insights into the pathogenesis and therapeutic targets for OPLL. LEVEL OF EVIDENCE: N/A.


Subject(s)
Cervical Vertebrae/metabolism , Genetic Predisposition to Disease/genetics , Hyperostosis/genetics , Hyperostosis/metabolism , Ossification of Posterior Longitudinal Ligament/genetics , Ossification of Posterior Longitudinal Ligament/metabolism , Aged , Animals , Cells, Cultured , Cervical Vertebrae/pathology , Female , Gene Expression , Genome-Wide Association Study/methods , Humans , Hyperostosis/pathology , Longitudinal Ligaments/metabolism , Longitudinal Ligaments/pathology , Male , Mice , Mice, Transgenic , Middle Aged , Ossification of Posterior Longitudinal Ligament/pathology
9.
FASEB J ; 34(7): 8876-8886, 2020 07.
Article in English | MEDLINE | ID: mdl-32510740

ABSTRACT

Autophagy, a macromolecular degradation process, plays a pivotal role in cell differentiation and survival. This study was designed to investigate the role of autophagy in the osteogenic differentiation in ligamentum fibroblasts. Rat ligamentum fibroblasts were isolated from the posterior longitudinal ligament and cultured in osteogenic induction medium. Ultrastructural analysis, immunofluorescence assay, western blot, flow cytometry, and lysosomal activity assessment were performed to determine the presence and activity of autophagy in the cells. The mineralization deposit and osteogenic gene expressions were evaluated to classify the association between autophagy activity and the bone formation ability of the spinal ligament cells. The influence of leptin and endothelin-1 on the autophagy activity was also evaluated. Our study demonstrated that autophagy was present and increased in the ligament cells under osteogenic induction. Inhibition of autophagy with either pharmacologic inhibitors (Bafilomycin A and 3-methyladenine) or Belcin1 (BECN1) knocking down weakened the mineralization capacity, decreased the gene expressions of COL1A1, osteocalcin (Ocn), and runt-related transcription factor 2 (Runx2) in the ligamentum fibroblasts and increased cell apoptosis. The Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK)-BECN1 autophagic pathway was activated in the osteogenic differentiating ligamentum fibroblasts. Leptin significantly increased the autophagy activity in the ligament cells under osteogenic induction. These discoveries might improve our understanding for the mechanism of ossification of the posterior longitudinal ligament (OPLL) and provide new approaches on the prevention and treatment of this not uncommon disease.


Subject(s)
Autophagy , Cell Differentiation , Fibroblasts/cytology , Longitudinal Ligaments/cytology , Ossification of Posterior Longitudinal Ligament/pathology , Osteogenesis , Animals , Cells, Cultured , Fibroblasts/metabolism , Longitudinal Ligaments/metabolism , Male , Ossification of Posterior Longitudinal Ligament/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
10.
Biochem J ; 477(12): 2249-2261, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32432317

ABSTRACT

Aberrant expression of microRNAs (miRNAs) has been associated with spinal ossification of the posterior longitudinal ligament (OPLL). Our initial bioinformatic analysis identified differentially expressed ADORA2A in OPLL and its regulatory miRNAs miR-497 and miR-195. Hence, this study was conducted to clarify the functional relevance of miR-497-195 cluster in OPLL, which may implicate in Adenosine A2A (ADORA2A). PLL tissues were collected from OPLL and non-OPLL patients, followed by quantification of miR-497, miR-195 and ADORA2A expression. The expression of miR-497, miR-195 and/or ADORA2A was altered in posterior longitudinal ligament (PLL) cells, which then were stimulated with cyclic mechanical stress (CMS). We validated that ADORA2A was expressed highly, while miR-497 and miR-195 were down-regulated in PLL tissues of OPLL patients. miR-195 and miR-497 expression in CMS-treated PLL cells was restored by a demethylation reagent 5-aza-2'-deoxycytidine (AZA). Moreover, expression of miR-195 and miR-497 was decreased by promoting promoter CpG island methylation. ADORA2A was verified as the target of miR-195 and miR-497. Overexpression of miR-195 and miR-497 diminished expression of osteogenic factors in PLL cells by inactivating the cAMP/PKA signaling pathway via down-regulation of ADORA2A. Collectively, miR-497-195 cluster augments osteogenic differentiation of PLL cells by inhibiting ADORA2A-dependent cAMP/PKA signaling pathway.


Subject(s)
Cell Differentiation , DNA Methylation , Gene Expression Regulation , MicroRNAs/genetics , Ossification of Posterior Longitudinal Ligament/pathology , Osteogenesis , Receptor, Adenosine A2A/metabolism , Adult , Aged , Female , Humans , Male , Middle Aged , Ossification of Posterior Longitudinal Ligament/genetics , Ossification of Posterior Longitudinal Ligament/metabolism , Receptor, Adenosine A2A/genetics , Signal Transduction
11.
Sci Rep ; 10(1): 7231, 2020 04 29.
Article in English | MEDLINE | ID: mdl-32350355

ABSTRACT

The pathomechanisms of initiation and progression of ossification of the posterior longitudinal ligament (OPLL) are unclear. Indian hedgehog (Ihh) and related signaling molecules are key factors in normal enchondral ossification. The purpose of this study is to investigate the contribution of mechanical strain to OPLL and the relationship of Ihh with OPLL. Sections of the posterior longitudinal ligament (PLL) were obtained from 49 patients with OPLL and from 7 patients without OPLL. Cultured PLL cells were subjected to 24 hours of cyclic tensile strain. To identify differentially expressed genes associated with cyclic tensile strain, microarray analysis was performed. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis identified upregulation of various genes, particularly of the Hedgehog signaling pathway; Ihh and related genes had increased expression compared with controls after 24-hour cyclic tensile strain. In immunoblotting analysis, Ihh, Runx2, Sox9, Gli2, Gli3, and smoothened (SMO) had significantly increased expression after 6- or 12-hour cyclic tensile strain. OPLL samples were strongly immunopositive for Ihh, Sox9, Runx2, Gli2, Gli3, and SMO in the ossification front of OPLL. These results suggest that cyclic tensile strain induces abnormal activation of Ihh and related signaling molecules, and this might be important in the ossification process in OPLL.


Subject(s)
Gene Expression Regulation , Hedgehog Proteins/metabolism , Longitudinal Ligaments/metabolism , Ossification of Posterior Longitudinal Ligament/metabolism , Signal Transduction , Stress, Mechanical , Aged , Female , Humans , Longitudinal Ligaments/pathology , Longitudinal Ligaments/surgery , Male , Middle Aged , Ossification of Posterior Longitudinal Ligament/pathology , Ossification of Posterior Longitudinal Ligament/surgery
12.
Mol Med Rep ; 21(1): 191-200, 2020 01.
Article in English | MEDLINE | ID: mdl-31939624

ABSTRACT

Thoracic ossification of the posterior longitudinal ligament (T­OPLL) is one of the most common factors that causes thoracic spinal stenosis, resulting in intractable myelopathy and radiculopathy. Our previous study reported that the rs201153092 polymorphism present in the collagen 6A1 (COL6A1) gene was a potentially pathogenic locus for the development of T­OPLL. The present study aimed to determine whether the rs201153092 mutation causes abnormal expression of COL6A1 in Han Chinese patients with T­OPLL, and to examine the effects of this mutation on osteogenesis by establishing a model of osteogenic differentiation. COL6A1 gene mutant and wild­type mouse 3T3­E1 embryonic osteoblast models were constructed to induce the differentiation of these cells into osteoblasts. The potential of the mutation site to induce abnormal expression of the COL6A1 gene and osteogenic markers was assessed via reverse transcription­quantitative PCR and western blot analyses. The results demonstrated that the rs201153092A mutation site resulted in significantly increased COL6A1 gene expression levels in the OPLL tissues obtained following clinical surgery. This mutation was shown to play an important role in the development of T­OPLL by regulating the overexpression of the COL6A1 gene and significantly increasing the expression levels of osteogenic markers. The findings of the present study suggested that the rs201153092A mutant variant could increase the expression levels of COL6A1 and consequently play a role in the pathogenesis of T­OPLL.


Subject(s)
Collagen Type VI/genetics , Longitudinal Ligaments/metabolism , Ossification of Posterior Longitudinal Ligament/genetics , Osteogenesis/genetics , Aged , Animals , Asian People , Cell Line , Collagen Type VI/metabolism , Female , Genetic Association Studies , Humans , Male , Mice , Middle Aged , Ossification of Posterior Longitudinal Ligament/metabolism , Ossification of Posterior Longitudinal Ligament/pathology , Osteoblasts , Polymorphism, Single Nucleotide , Thoracic Vertebrae/metabolism , Thoracic Vertebrae/pathology
13.
FEBS Lett ; 592(24): 4051-4065, 2018 12.
Article in English | MEDLINE | ID: mdl-30381822

ABSTRACT

Through their multiple targets, microRNAs (miRNAs) are involved in numerous physiological and pathological processes. In this study, miR-342-3p was found to be deregulated with ossification of ligament or osteoporosis. We demonstrate that silencing miR-342-3p impairs osteoblast activity and matrix mineralization, while over expression of miR-342-3p promotes osteoblast differentiation significantly. Moreover, miR-342-3p directly targets activating transcription factor 3 (ATF3), which inhibits transcription of pro-osteogenic differentiation-associated genes. In addition, there exists a higher frequency of methylation at the CpG island of the Enah/Vasp-Like (EVL) locus in undifferentiated pre-osteoblasts; however, demethylation of the EVL CpG island induces over expression of miR-342-3p during osteogenic differentiation. This study suggests that miR-342-3p may serves as a potential marker for diagnosis and treatment of ossification of ligament and osteoporosis.


Subject(s)
Activating Transcription Factor 3/genetics , Cell Differentiation/genetics , MicroRNAs/genetics , Osteoblasts/metabolism , Osteogenesis/genetics , Activating Transcription Factor 3/metabolism , Animals , Cell Line , Cells, Cultured , Female , Gene Expression Profiling , HEK293 Cells , HT29 Cells , Humans , Male , Mice, Inbred C57BL , Ossification of Posterior Longitudinal Ligament/genetics , Ossification of Posterior Longitudinal Ligament/metabolism , Osteoblasts/cytology , Osteoporosis/genetics , Osteoporosis/metabolism
14.
J Orthop Surg Res ; 13(1): 165, 2018 Jul 03.
Article in English | MEDLINE | ID: mdl-29970120

ABSTRACT

BACKGROUND: Hyperleptinemia is a common feature of obese people, and leptin, an adipocyte-derived cytokine, is believed to be an important factor in the pathogenesis of cervical ossification of the posterior longitudinal ligament(C-OPLL). So this research was to identify the relation between the serum leptin and bone metabolic markers and how the leptin induced osteogenic effect in C-OPLL. METHODS: Sixty-four samples were selected to determine the concentration of leptin, insulin, and alkaline phosphatase. And the association of leptin with these factors was also examined. We also evaluate the effect of leptin on the development of C-OPLL and further explored the possible underlying mechanism in vitro. RESULTS: We found that serum leptin concentrations were higher in females than in males. Serum leptin and ALP concentrations were increased significantly in C-OPLL females compared to non-OPLL females. In OPLL subjects, the serum leptin concentration corrected for body mass index correlated negatively with the ALP concentrations. In C-OPLL cells, leptin treatment led to a significant increase in mRNA expressions of ALP and OCN and formation of mineralized nodule. Our experiments reported here that osteogenic effect of leptin in C-OPLL cells could be mediated via ERK1/2, p38 MAPK, and/or JNK signaling pathways. CONCLUSIONS: From this research, we got that leptin treatment led to a significant increase in mRNA expressions of ALP and OCN and formation of mineralized nodule. And the osteogenic effect of leptin in C-OPLL cells could be mediated via ERK1/2, p38 MAPK, and/or JNK signaling pathways.


Subject(s)
Leptin/metabolism , Longitudinal Ligaments/metabolism , Ossification of Posterior Longitudinal Ligament/metabolism , Ossification, Heterotopic/metabolism , Aged , Alkaline Phosphatase/blood , Female , Humans , Insulin/blood , Leptin/blood , Leptin/pharmacology , Longitudinal Ligaments/drug effects , Male , Middle Aged , Neck , Ossification of Posterior Longitudinal Ligament/blood , Ossification, Heterotopic/blood
15.
Eur Spine J ; 27(8): 1757-1766, 2018 08.
Article in English | MEDLINE | ID: mdl-29909551

ABSTRACT

PURPOSE: To evaluate the effects of leptin/leptin receptor (LepR) combined with mechanical stress on the development of ossification of the posterior longitudinal ligament (OPLL), which is a disease characterized by ectopic bone formation of the posterior longitudinal ligament (PLL) and can lead to radiculopathy and myelopathy. METHODS: Six human samples of the PLL were analyzed for the expression of leptin and LepR by RT-PCR and western blotting. PLL cells were stimulated with leptin and mechanical stress delivered via a Flexcell tension system, and osteogenic differentiation was evaluated by RT-PCR and western blotting analysis of osteogenic marker expression as well as by alkaline phosphatase (ALP) staining and alizarin red S staining. Activation of mitogen-activated protein kinase (MAPK), Janus kinase (JAK) 2-signal transducer, activator of transcription (STAT) 3 and phosphatidylinositol 3-kinase (PI3K)-Akt was evaluated by western blotting. RESULTS: Samples from the OPLL group had higher LepR mRNA and protein levels and lower leptin levels than those from healthy controls. Exposure to leptin and Flexcell increased the number of ALP-positive cells and calcium nodules in a dose-dependent manner; this effect was accompanied by upregulation of the osteogenic markers osteocalcin, runt-related transcription factor 2 (RUNX2) and osteopontin. Extracellular signal-regulated kinase, P38 MAPK, JAK2, STAT3, PI3K and Akt signaling, was also activated by the combined effects of leptin and mechanical stress. CONCLUSIONS: Leptin and LepR are differentially expressed in OPLL tissues, and the combined use of leptin/LepR and mechanical stress promotes osteogenic differentiation of PLL cells via MAPK, JAK2-STAT3 and PI3K/Akt signaling. These slides can be retrieved under Electronic Supplementary Material.


Subject(s)
Leptin/metabolism , Ossification of Posterior Longitudinal Ligament/metabolism , Ossification, Heterotopic/metabolism , Receptors, Leptin/metabolism , Stress, Mechanical , Alkaline Phosphatase/metabolism , Blotting, Western , Cell Culture Techniques , Cell Differentiation , Humans , Longitudinal Ligaments/cytology , Longitudinal Ligaments/metabolism , Longitudinal Ligaments/pathology , Ossification of Posterior Longitudinal Ligament/etiology , Ossification, Heterotopic/etiology , Real-Time Polymerase Chain Reaction , Signal Transduction
16.
Sci Rep ; 8(1): 9225, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29907859

ABSTRACT

Ossification of the posterior longitudinal ligament (OPLL) presents as pathological heterotopic ossification of the spinal ligaments. However, its underlying molecular mechanism is still unclear. Our previous findings suggested that altered microRNA regulatory network are critical for the development of OPLL. Here, we set out to unveiling the detailed mechanism of those altered OPLL-specific microRNAs. We screened a set of differentially expressed OPLL-specific microRNAs from the previous sequencing data and showed that microRNA-10a actively modulates the ossification of posterior ligament cells in vitro. Using a tissue-engineered scaffold grown from 4-week-old BALB/c homozygous nude mice, we found that altered microRNA-10a expression in posterior ligament cells indeed affected the heterotopic bone formation in vivo. Furthermore, computational analysis showed that the negative ossification regulator ID3 is a functional target gene of microRNA-10a, and its expression was also significantly altered during microRNA-10a modulation both in vitro and in vivo. Also, we have demonstrated that the ossification promoting function of microRNA-10a requires ID3, as ID3 actively inhibits RUNX2. Thus, we identified a critical role for highly altered OPLL-specific microRNA-10a in regulating the development of OPLL by modulating the ID3/RUNX2 axis.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Inhibitor of Differentiation Proteins/metabolism , Longitudinal Ligaments/metabolism , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Ossification of Posterior Longitudinal Ligament/metabolism , Ossification, Heterotopic/metabolism , Animals , Cells, Cultured , Female , Humans , Longitudinal Ligaments/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Ossification of Posterior Longitudinal Ligament/pathology , Ossification, Heterotopic/pathology
17.
PLoS One ; 13(5): e0196204, 2018.
Article in English | MEDLINE | ID: mdl-29782494

ABSTRACT

Ossification of the posterior longitudinal ligament (OPLL), a spinal ligament, reduces the range of motion in limbs. No treatment is currently available for OPLL, which is why therapies are urgently needed. OPLL occurs in obesity, is more common in men, and has an onset after 40 years of age. The mechanisms underlying OPLL remain unclear. In this study, we performed a serum proteomic analysis in both OPLL patients and healthy subjects to identify factors potentially involved in the development of OPLL, and found reduced levels of a protein that might underlie the pathology of OPLL. We isolated the protein, determined its amino acid sequence, and identified it as chemokine (C-X-C motif) ligand 7 (CXCL7). Based on these proteomics findings, we generated a CXCL7 knockout mouse model to study the molecular mechanisms underlying OPLL. CXCL7-null mice presented with a phenotype of OPLL, showing motor impairment, heterotopic ossification in the posterior ligament tissue, and osteoporosis in vertebrate tissue. To identify the mechanisms of CXCL7 deficiency in OPLL, we searched for single nucleotide polymorphisms and altered DNA exons, but no abnormalities were found. Although miR-340 levels were found to be high in an miRNA array, they were insufficient to reduce CXCL7 levels. Ubiquitin C-terminal hydrolase1 (UCHL1) was found to be overexpressed in CXCL7-null mice and in the sera of patients with OPLL, and was correlated with OPLL severity. Post-translational modifications of proteins with ubiquitin and ubiquitin-like modifiers, orchestrated by a cascade of specialized ubiquitin activating enzyme (E1), ubiquitin conjugating enzyme (E2), and ubiquitin ligase (E3) enzymes, are thought to control a wide range of cellular processes, and alterations in the ubiquitin-proteasome system have been associated with several degenerative disorders. In addition, the OPLL tissue of CXCL7-null mouse and its primary cells expressed the antibody to ubiquitin (linkage-specific K48). Our data clearly show decreased CXCL7 levels in patients with OPLL, and that OPLL developed in mice lacking CXCL7. Tumor necrosis factor receptor-associated factor (TRAF)6 expression was decreased in CXCL7-null mouse primary cells. Furthermore, K48 polyubiquitination was found in posterior longitudinal ligament ossified tissue and primary cells from CXCL7-null mice. We performed a phosphoproteomics analysis in CXCL7-deficient mice and identified increased phosphorylation of mitogen-activated protein kinase kinase (ME3K)15, ubiquitin protein ligase E3C (UBE3C) and protein kinase C (PKC) alpha, suggesting that ubiquitin-dependent degradation is involved in CXCL7 deficiency. Future studies in the CXCL7-null mouse model are, therefore, warranted to investigate the role of ubiquitination in the onset of OPLL. In conclusion, CXCL7 levels may be useful as a serum marker for the progression of OPLL. This study also suggests that increasing CXCL7 levels in patients can serve as an effective therapeutic strategy for the treatment of OPLL.


Subject(s)
Biomarkers/metabolism , Chemokines, CXC/physiology , Ossification of Posterior Longitudinal Ligament/pathology , Ubiquitin-Protein Ligases/metabolism , beta-Thromboglobulin/metabolism , Aged , Animals , Female , Humans , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Middle Aged , Ossification of Posterior Longitudinal Ligament/genetics , Ossification of Posterior Longitudinal Ligament/metabolism , Phenotype , Proteolysis , Proteomics , Ubiquitins/metabolism , beta-Thromboglobulin/genetics
18.
Exp Cell Res ; 349(1): 53-59, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27693496

ABSTRACT

Ossification of the posterior longitudinal ligament (OPLL) involves ectopic calcification of the spinal ligament preferentially at the cervical spine. OPLL is associated with different diseases and occurs by endochondral ossification, which is associated with the activity of different transcription factors. However, the pathogenesis of OPLL remains unclear. Here, we investigated the role of osterix (Osx), a transcription factor that functions downstream of Runx2 and is an important regulator of osteogenesis, in the process of OPLL in a dexamethasone (Dex)-induced model of spinal ligament ossification. Our results showed that Osx is upregulated in patients with OPLL and during the ossification of ligament cells in parallel with the upregulation of osteogenic markers including osteocalcin (OCN), alkaline phosphatase (ALP) and collagen-1 (Col-1). Dex-induced ossification of ligament cells was associated with the downregulation and inactivation of ß-catenin, and these effects were offset by Osx knockdown. Activation of ß-catenin signaling abolished the effect of Dex on ossification and the upregulation of osteogenic markers. Taken together, our results suggest that OPLL is mediated by Osx via a mechanism involving the Wnt/ß-catenin signaling pathway, providing a basis for further research to identify potential targets for the treatment of OPLL.


Subject(s)
Ossification of Posterior Longitudinal Ligament/metabolism , Signal Transduction , Transcription Factors/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing , Bone Morphogenetic Proteins/metabolism , Dexamethasone/pharmacology , Down-Regulation/drug effects , Gene Silencing/drug effects , Genetic Markers , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lithium Chloride/pharmacology , Longitudinal Ligaments/metabolism , Longitudinal Ligaments/pathology , Ossification of Posterior Longitudinal Ligament/pathology , Signal Transduction/drug effects , Sp7 Transcription Factor , Up-Regulation/drug effects , Wnt Proteins/metabolism
19.
Sci Rep ; 6: 26962, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27246988

ABSTRACT

Ossification of the posterior longitudinal ligament of the spine (OPLL), which is characterized by ectopic bone formation in the spinal ligaments, can cause spinal-cord compression. To date, at least 11 susceptibility genes have been genetically linked to OPLL. In order to identify potential deleterious alleles in these OPLL-associated genes, we designed a capture array encompassing all coding regions of the target genes for next-generation sequencing (NGS) in a cohort of 55 unrelated patients with OPLL. By bioinformatics analyses, we successfully identified three novel and five extremely rare variants (MAF < 0.005). These variants were predicted to be deleterious by commonly used various algorithms, thereby resulting in missense mutations in four OPLL-associated genes (i.e., COL6A1, COL11A2, FGFR1, and BMP2). Furthermore, potential effects of the patient with p.Q89E of BMP2 were confirmed by a markedly increased BMP2 level in peripheral blood samples. Notably, seven of the variants were found to be associated with the patients with continuous subtype changes by cervical spinal radiological analyses. Taken together, our findings revealed for the first time that deleterious coding variants of the four OPLL-associated genes are potentially pathogenic in the patients with OPLL.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Collagen Type VI/genetics , Collagen Type XI/genetics , Genetic Predisposition to Disease , Mutation, Missense , Ossification of Posterior Longitudinal Ligament/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Adult , Aged , Bone Morphogenetic Protein 2/metabolism , Cohort Studies , Collagen Type VI/metabolism , Collagen Type XI/metabolism , Computational Biology , Female , Gene Expression , Genetic Association Studies , High-Throughput Nucleotide Sequencing , Humans , Longitudinal Ligaments/diagnostic imaging , Longitudinal Ligaments/metabolism , Longitudinal Ligaments/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Ossification of Posterior Longitudinal Ligament/diagnostic imaging , Ossification of Posterior Longitudinal Ligament/metabolism , Ossification of Posterior Longitudinal Ligament/pathology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Spine/diagnostic imaging , Spine/metabolism , Spine/pathology , Tomography, X-Ray Computed
20.
Sci Rep ; 6: 21580, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26868491

ABSTRACT

Ossification of the posterior longitudinal ligament (OPLL) is a genetic disorder which involves pathological heterotopic ossification of the spinal ligaments. Although studies have identified several genes that correlated with OPLL, the underlying regulation network is far from clear. Through small RNA sequencing, we compared the microRNA expressions of primary posterior longitudinal ligament cells form OPLL patients with normal patients (PLL) and identified 218 dysregulated miRNAs (FDR < 0.01). Furthermore, assessing the miRNA profiling data of multiple cell types, we found these dysregulated miRNAs were mostly OPLL specific. In order to decipher the regulation network of these OPLL specific miRNAs, we integrated mRNA expression profiling data with miRNA sequencing data. Through computational approaches, we showed the pivotal roles of these OPLL specific miRNAs in heterotopic ossification of longitudinal ligament by discovering highly correlated miRNA/mRNA pairs that associated with skeletal system development, collagen fibril organization, and extracellular matrix organization. The results of which provide strong evidence that the miRNA regulatory networks we established may indeed play vital roles in OPLL onset and progression. To date, this is the first systematic analysis of the micronome in OPLL, and thus may provide valuable resources in finding novel treatment and diagnostic targets of OPLL.


Subject(s)
Gene Expression Regulation , Longitudinal Ligaments/metabolism , MicroRNAs/biosynthesis , Ossification of Posterior Longitudinal Ligament/metabolism , Cells, Cultured , Female , High-Throughput Nucleotide Sequencing , Humans , Longitudinal Ligaments/pathology , Male , Ossification of Posterior Longitudinal Ligament/diagnosis , Ossification of Posterior Longitudinal Ligament/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...