Mechanistic Roles of Leptin in Osteogenic Stimulation in Thoracic Ligament Flavum Cells
Dongwei Fan,Zhongqiang Chen,Yupeng Chen,Yongfeng Shang
DOI: https://doi.org/10.1074/jbc.m611779200
2007-01-01
Abstract:Obesity is a risk factor for thoracic ossification of ligament flavum (TOLF) that is characterized by ectopic bone formation in the spinal ligaments. Hyperleptinemia is a common feature of obese people, and leptin, an adipocyte-derived cytokine with proliferative and osteogenic effects in several cell types, is believed to be an important factor in the pathogenesis of TOLF. However, how leptin might stimulate cell osteogenic differentiation in TOLF is not totally understood. We reported here that leptin-induced osteogenic effect in TOLF cells is associated with activation of signaling molecules STAT3, JNK, and ERK1/2 but not p38. Blocking STAT3 phosphorylation with a selective inhibitor, AG490, significantly abolished leptin-induced osteogenic differentiation of TOLF cells, whereas blocking ERK1/2 and JNK phosphorylation with their selective inhibitors PD98059 and SP600125, respectively, had only marginal effects. In addition, we showed that STAT3 interacted with Runt-related transcription factor 2 (Runx2) in the nucleus, and STAT3, Runx2, and steroid receptor coactivator steroid receptor coactivator-1 were components of the transcription complex recruited on Runx2 target gene promoters in response to leptin treatment. Our experiments identified STAT3, Runx2, and steroid receptor coactivator-1 as critical molecules in mediating leptin-stimulated cell osteogenesis in TOLF. Obesity is a risk factor for thoracic ossification of ligament flavum (TOLF) that is characterized by ectopic bone formation in the spinal ligaments. Hyperleptinemia is a common feature of obese people, and leptin, an adipocyte-derived cytokine with proliferative and osteogenic effects in several cell types, is believed to be an important factor in the pathogenesis of TOLF. However, how leptin might stimulate cell osteogenic differentiation in TOLF is not totally understood. We reported here that leptin-induced osteogenic effect in TOLF cells is associated with activation of signaling molecules STAT3, JNK, and ERK1/2 but not p38. Blocking STAT3 phosphorylation with a selective inhibitor, AG490, significantly abolished leptin-induced osteogenic differentiation of TOLF cells, whereas blocking ERK1/2 and JNK phosphorylation with their selective inhibitors PD98059 and SP600125, respectively, had only marginal effects. In addition, we showed that STAT3 interacted with Runt-related transcription factor 2 (Runx2) in the nucleus, and STAT3, Runx2, and steroid receptor coactivator steroid receptor coactivator-1 were components of the transcription complex recruited on Runx2 target gene promoters in response to leptin treatment. Our experiments identified STAT3, Runx2, and steroid receptor coactivator-1 as critical molecules in mediating leptin-stimulated cell osteogenesis in TOLF. Ossification of ligament flavum (OLF) 3The abbreviations used are: OLF, ossification of ligament flavum; TOLF, thoracic OLF; SRC, steroid receptor coactivator; Runx2, Runt-related transcription factor 2; STAT3, signal transducer and activator of transcription 3; MAPK, mitogen-activated protein kinase; ALP, alkaline phosphatase activity; ChIP, chromatin Immunoprecipitation; Re-ChIP, ChIP re-immunoprecipitation; JNK, c-Jun NH2-terminal kinase; ERK, extracellular signal-regulated kinase; DMEM, Dulbecco's modified Eagle's medium; FBS, fetal bovine serum; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; RT, reverse transcription. of the spine is characterized by a heterotopic bone formation in the ligament flavum that is normally composed of fibrous tissues (1al-Orainy I.A. Kolawole T. Eur. J. Radiol. 1998; 29: 76-82Abstract Full Text Full Text PDF PubMed Scopus (38) Google Scholar). Ossification could enlarge the spinal canal and compresses the spinal cord, resulting in serious neurological damages. Epidemiology has shown that high incidence rate of OLF occurs in thoracic spine (2Ben Hamouda K. Jemel H. Haouet S. Khaldi M. J. Neurosurg. 2003; 99: 157-161PubMed Google Scholar). It has been documented that obesity represents a risk factor for thoracic ossification of ligament flavum (TOLF), particularly in Asian people (3Vasudevan A. Knuckey N.W. J. Clin. Neurosci. 2002; 9: 311-313Abstract Full Text PDF PubMed Scopus (22) Google Scholar). Indeed, hereditary obese rats, Zucker fatty (fa/fa) rats, are prone to OLF (4Okano T. Ishidou Y. Kato M. Imamura T. Yonemori K. Origuchi N. Matsunaga S. Yoshida H. ten Dijke P. Sakou T. J. Orthop. Res. 1997; 15: 820-829Crossref PubMed Scopus (20) Google Scholar). A common feature of obese people is hyperleptinemia (5Shirakura Y. Sugiyama T. Tanaka H. Taguchi T. Kawai S. Biochem. Biophys. Res. Commun. 2000; 267: 752-755Crossref PubMed Scopus (26) Google Scholar). Leptin, an adipocyte-derived cytokine, can stimulate the proliferation and osteogenic differentiation of various cell lines, such as the embryonic cell line C3H10T1/2, human NHOst cells, and human osteoblastic cells (6Hess R. Pino A.M. Rios S. Fernandez M. Rodriguez J.P. J. Cell. Biochem. 2005; 94: 50-57Crossref PubMed Scopus (69) Google Scholar, 7Iwamoto I. Fujino T. Douchi T. Gynecol. Endocrinol. 2004; 19: 97-104Crossref PubMed Scopus (44) Google Scholar). However, the molecular mechanism underlying the osteogenic effect of leptin in TOLF is not totally understood. Leptin exerts its biological activity through binding to its receptors, which belong to cytokine receptor superfamily. Different leptin receptor isoforms exist, including a long form (ObRb) and a short form (ObRa) (8Aleffi S. Petrai I. Bertolani C. Parola M. Colombatto S. Novo E. Vizzutti F. Anania F.A. Milani S. Rombouts K. Laffi G. Pinzani M. Marra F. Hepatology. 2005; 42: 1339-1348Crossref PubMed Scopus (309) Google Scholar). In vitro and in vivo studies have shown that leptin activates cytokine-like signal transduction via the long form receptor. Upon leptin stimulation, intracellular Janus tyrosine kinases are activated via transphosphorylation and phosphorylate tyrosine residues on the long form leptin receptor and on signal transducers and activators of transcription (STAT) proteins (9Zhang F. Chen Y. Heiman M. Dimarchi R. Vitam. Horm. 2005; 71: 345-372Crossref PubMed Scopus (253) Google Scholar). Phosphorylated STAT proteins dimerize and translocate to the nucleus to activate gene transcription. We report here that STAT3 signaling pathway was involved in the osteogenic differentiation of TOLF cells in response to leptin. The p160 family of nuclear receptor coactivators SRC-1, but not GRIP1 and AIB1, interacted with STAT3. We showed that STAT3, SRC-1, and Runx2 are components of the transcription complex recruited to the Runx2 target gene promoters to regulate the target gene transcription. Antibodies and Reagents—Polyclonal anti-ob-receptor, polyclonal anti-ERK1/2, anti-p38 MAPK, anti-JNK, anti-phospho-ERK1/2, anti-phospho-p38 MAPK, anti-phospho-JNK, anti-osteocalcin, and anti-Runx2 were obtained from Santa Cruz Biotechnology (Santa Cruz, CA). Polyclonal anti-STAT3 and anti-phospho-STAT3 Tyr705 were from Upstate Biotechnology (Lake Placid, NY) and New England Biolabs (Beverly, MA), respectively. Recombinant human leptin was from R&D Systems (Minneapolis, MN). PD98059, SP600125, and AG490 were obtained from Promega and Calbiochem and were dissolved in dimethyl sulfoxide (Me2SO) before use. Clinical Diagnosis and Spinal Ligament Samples—The diagnosis of TOLF or non-TOLF (i.e. other thoracic diseases) was confirmed by x-ray, computerized tomography, and magnetic resonance imaging of the whole spine preoperatively to avoid any other non-thoracic OLF patients. The clinical diagnoses and the spinal ligament tissues used in this study are shown in Table 1. Ligaments were aseptically harvested from patients during surgery and rinsed with phosphate-buffered saline. Surrounding tissue was carefully removed under a dissecting microscope. In all cases the ligaments were extirpated carefully from nonossified sites to avoid any possible contamination of osteogenic cells. This study was approved by the Ethics Committee of Peking University Health Science Center.TABLE 1Tissue samples used in the study, showing clinical diagnosis, patient's sex and age, and tissue derived TT, thoracic trauma; ST, spinal tuberculosis; TSS, thoracolumber spinal stenosis; TF, thoracic fibroneuroma; M, male; F, female; BMI, body mass index.TOLFNon-TOLFCodeDiagnosisSex/ageTissueBMICodeDiagnosisSex/ageTissueBMIO-1aMean obesityTOLFM/51TLF25.6N-1TTM/46TLF23.7O-2TOLFM/52TLF23.1N-2STM/48TLF23.2O-3aMean obesityTOLFM/60TLF25.3N-3TTM/47TLF23.4O-4aMean obesityTOLFM/68TLF25.7N-4aMean obesityTTM/56TLF25.3O-5TOLFM/69TLF23.3N-5TSSM/57TLF22.8O-6aMean obesityTOLFF/48TLF26.2N-6TTF/43TLF23.5O-7aMean obesityTOLFF/53TLF25.5N-7aMean obesityTTF/57TLF25.2O-8aMean obesityTOLFF/64TLF26.1N-8aMean obesityTTF/65TLF25.1O-9aMean obesityTOLFF/65TLF25.8N-9TFF/66TLF23.4O-10aMean obesityTOLFF/66TLF25.1N-10TTF/73TLF23.1a Mean obesity Open table in a new tab Cell Cultures—Collected ligaments were minced into about 0.5-mm3 pieces and washed twice with phosphate-buffered saline then plated in 6-cm culture dishes and maintained in DMEM medium (10% FBS, 1% l-glutamine, 100 units/ml of penicillin G sodium, 100 μg/ml of streptomycin sulfate) in a humidified atmosphere of 95% air and 5% CO2 at 37 °C. The cells derived from explants were removed from the dish with 0.02% EDTA, 0.05% trypsin for passage. The first and third passage cells were used in the following studies (10Specchia N. Pagnotta A. Gigante A. Logroscino G. Toesca A. J. Orthop. Res. 2001; 19: 294-300Crossref PubMed Scopus (41) Google Scholar). Colorimetric 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium Bromide (MTT) Assay—Cell proliferation was measured by MTT dye reduction assay. Briefly, cells were seeded into 96-well plates overnight and exposed to the leptin at different concentrations for different times. The cells then were incubated with MTT (0.5 mg/ml) for 4 h at 37 °C. After removal of MTT, 150 μl of Me2SO were added to the cells, and the plates were shaken at room temperature for 10 min. The absorbance was measured at 540 nm using a microplate reader (7Iwamoto I. Fujino T. Douchi T. Gynecol. Endocrinol. 2004; 19: 97-104Crossref PubMed Scopus (44) Google Scholar). Mineralization Assay—Cells from TOLF patients (TOLF cells) and non-TOLF patients (non-TOLF cells) were plated at 40,000 cells/well in 6-well dishes and maintained in DMEM supplemented with 10% FBS. On confluence, designated day 0, cells were exposed to leptin medium containing DMEM supplemented with 10% FBS, 10 mm β-glycerophosphate, 100 ng/ml leptin. Medium was replaced for every 3 to 4 days. Samples were processed at 4 weeks, and alizarin red assay (Sigma) was performed to determine mineralization. Briefly, cells were washed with phosphate-buffered saline and water and fixed with ice-cold 100% ethanol for 60 min at 4 °C. Fixed cultures were incubated with 1% alizarin red for 30 min and washed with distilled water for several times. Extracellular matrix mineral-bound stain was photographed under microscopy (11Huitema L.F. Vaandrager A.B. van Weeren P.R. Barneveld A. Helms J.B. van de Lest C.H. Calcif. Tissue Int. 2006; 78: 171-177Crossref PubMed Scopus (6) Google Scholar). Semiquantitative PCR and Real-time PCR—Total RNA was extracted from the cell monolayers using TRIzol reagents (Invitrogen), and any potential DNA contamination was removed by RNase-free DNase treatment. mRNA expression of various genes was determined by reverse transcription (RT)-PCR. Two micrograms of total RNA were reverse-transcribed using the Superscript first-strand synthesis system for RT-PCR (Invitrogen). Primer sets used for amplification were: osteocalcin, 5′-AGGGCAGCGAGGTAGTGA-3′ (forward), 5′-CCTGAAAGCCGATGTGGT-3′ (reverse); ALP, 5′-CTGATGTGGAGTATGA-3′ (forward), 5′-TGTATCTCGGTTTGAA-3′ (reverse); β-actin 5′-TTAGTTGCGTTACACCCTTTC-3′ (forward), 5′-GCTGT CACCT TCACC GTTC-3′ (reverse); ObRb, 5′-TCACCCAGTGATTACAAGCT-3′ (forward), 5′-CTGGAGAACTCTGATGTCCG-3′ (reverse). For quantification of gene expression, all products were assayed in the exponential phase of the amplification curve, and the PCR cycles were determined for each primer pair. Amplified products were separated on 1.5% agarose gels and stained with ethidium bromide for visualization. Visualized PCR product bands were detected by AlphaImager 2200 (Alpha Innotech). All products were normalized to β-actin mRNA levels. For real-time PCR, PCR reactions were performed in triplicates using the SYBR® Green Master Mix (ABI, Warrington, UK) and the ABI 7300 Real Time PCR system. Transcript levels were normalized to β-actin levels. ALP Assays—The ALP activity was measured after rinsing the cells twice with ice-cold phosphate-buffered saline (pH 7.4), then solubilized in Tris/glycine/Triton buffer (pH 8.5, 50 mm Tris, 100 mm glycine, and 0.1% Triton X-100) and sonicated on ice. This mixture was centrifuged at 5000 × g for 15 min at 4 °C, and the supernatant was collected. One hundred microliters of freshly prepared p-nitrophenyl-phosphate substrate (1.5 mg/ml) was added to 200 μl of the supernatant and incubated for 30 min at 37 °C. For the generation of a standard curve, serial dilutions of a p-nitrophenol standard solution were prepared, and 100 μl of each concentration was included in each tube. The enzymatic reaction was terminated with the addition of 0.3 ml of ice-cold NaOH (0.1 m) solution. The absorbance was read at 405 nm. Concentrations of protein were determined with a Bradford protein assay using BSA as standard. Results are expressed as nmol of p-nitrophenol/μg of cellular protein/min (12Ciancaglini P. Simao A.M. Camolezi F.L. Millan J.L. Pizauro J.M. Braz. J. Med. Biol. Res. 2006; 39: 603-610Crossref PubMed Scopus (40) Google Scholar). Luciferase Assays—Cells were transfected with an osteocalcin promoter-driven luciferase construct (pOC-LUC) (13Sierra J. Villagra A. Paredes R. Cruzat F. Gutierrez S. Javed A. Arriagada G. Olate J. Imschenetzky M. Van Wijnen A.J. Lian J.B. Stein G.S. Stein J.L. Montecino M. Mol. Cell. Biol. 2003; 23: 3339-3351Crossref PubMed Scopus (189) Google Scholar) using Lipofectamine 2000 (Invitrogen) according to the manufacturer's instruction. Forty-eight hours after transfection, the cells were harvested, and luciferase and renilla activities were measured using dual luciferase kit (Promega). The firefly luciferase data for each sample were normalized based on transfection efficiency measured by renilla luciferase activity. Each assay was performed in triplicate and repeated at least three times. Immunoprecipitation and Western Blotting—Nuclear extracts were precleaned with 50 μl of protein A-agarose beads for 30 min followed by pelleting the beads. Anti-Runx2 antibody was then added and incubated for 2 h at 4 °C with gentle rocking. The immune complexes were collected after the addition of 30 μl of protein A-agarose beads and incubation for 1 h at 4 °C followed by centrifugation. Precipitates were suspended in washing buffer (20 mm Tris-HCl (pH 8.3), 0.5% sodium deoxycholate, 0.5% Nonidet P-40, 50 mm NaCl, 2 mm EDTA, and 0.2 mm phenylmethylsulfonyl fluoride), centrifuged, and resuspended in 0.5 m Tris-buffered saline (pH 6.8), containing 2% SDS, 1 mm dithiothreitol, 10% glycerol, and 0.01% phenol blue. The protein samples were then denatured at 95 °C for 4 min, resolved by 10% SDS-PAGE, and transblotted to Hybond/ECL nitrocellulose membranes. The membranes were blocked overnight in Tris-buffered saline containing 1% bovine serum albumin, 1% polyvinylpyrrolidone, and 0.01% Tween 20 and incubated with an appropriately diluted primary antibody followed by incubation with horseradish peroxidase-conjugated secondary IgG. Signals were visualized with diaminobenzidine on LAS3000 Lumi-Imager (Fuji Photo Film Co., Ltd.). Chromatin Immunoprecipitation (ChIP) and ChIP Re-immunoprecipitation (Re-ChIP) Assay—ChIP experiments were performed according to the method described elsewhere (14Shang Y. Hu X. DiRenzo J. Lazar M.A. Brown M. Cell. 2000; 103: 843-852Abstract Full Text Full Text PDF PubMed Scopus (1469) Google Scholar, 15Zhang H. Yi X. Sun X. Yin N. Shi B. Wu H. Wang D. Wu G. Shang Y. Genes Dev. 2004; 18: 1753-1765Crossref PubMed Scopus (82) Google Scholar, 16Yin N. Wang D. Zhang H. Yi X. Sun X. Shi B. Wu H. Wu G. Wang X. Shang Y. Cancer Res. 2004; 64: 5870-5875Crossref PubMed Scopus (174) Google Scholar, 17Wu H. Chen Y. Liang J. Shi B. Wu G. Zhang Y. Wang D. Li R. Yi X. Zhang H. Sun L. Shang Y. Nature. 2005; 438: 981-987Crossref PubMed Scopus (241) Google Scholar, 18Zhang H. Sun L. Liang J. Yu W. Zhang Y. Wang Y. Chen Y. Li R. Sun X. Shang Y. EMBO J. 2006; 25: 4223-4233Crossref PubMed Scopus (72) Google Scholar). The presence of the target gene promoter sequences in both the input DNA and the recovered DNA immunocomplexes were detected by real-time PCR using DNA as template for real-time PCR with SYBR Green PCR Master Mix (Applied Biosystems). The primer sequences for specific promoter regions (within -500 to +100 regions, corresponding to the transcription start sites of genes) are shown below. The data obtained were normalized to the corresponding DNA input control. The sequences of the primers used were as follows: for osteocalcin: forward, 5′-CGGGCAGTCTGATTGTGGC-3′ (position -261), and reverse, 5′-CTCATGGTGTCTCGGTGGC-3′ (position +23); control sequence, forward, 5′-AAAGTGGTGGTGACAAAGGC-3′ (position -3368), and reverse, 5′-TGAGTGGCGATAGTGGAGC-3′ (position -3234). The Re-ChIP assays were performed as described earlier (15Zhang H. Yi X. Sun X. Yin N. Shi B. Wu H. Wang D. Wu G. Shang Y. Genes Dev. 2004; 18: 1753-1765Crossref PubMed Scopus (82) Google Scholar). The immunoprecipitated complexes obtained by ChIP were eluted by incubation for 30 min at 37 °C in 25 μl of 10 mm dithiothreitol. After centrifugation, the supernatant was diluted 20 times with sonication buffer and subjected to the ChIP procedure. Statistical Analysis—The data were analyzed by analysis of variance, and the Student-Newman-Kleuss method was used to estimate the level of significance. p < 0.05 was considered significant. Effect of Leptin on Proliferation and Osteogenic Differentiation of TOLF and Non-TOLF Cells—As stated above, hyperleptinemia is a risk factor for TOLF; to investigate the molecular mechanism underlying leptin-stimulated TOLF, we first examined the effect of leptin on the proliferation and osteogenic differentiation of TOLF and non-TOLF cells. In these experiments TOLF and non-TOLF cells were treated with various concentrations of leptin for different periods of time. The effect of leptin treatment on cell proliferation was evaluated by MTT methodologies, and cell osteogenic differentiation was assessed by real-time RT-PCR, Western blotting, ALP assay, and mineralization assay. As shown in Fig. 1, real-time RT-PCR analysis showed that whereas leptin stimulated the proliferation of a breast carcinoma cell line, MCF-7, it had no significant effect on the proliferation of either TOLF cells or non-TOLF cells (Fig. 1A). However, leptin treatment resulted in a significant increase in mRNA expression of ALP and osteocalcin in TOLF cells but not in non-TOLF cells, and the effect was both dose- and time-dependent (Fig. 1, B–E). In agreement with the real-time RT-PCR experiments, ALP activity assay and Western blotting demonstrated that both the activity of ALP and the protein expression of osteocalcin were elevated in response to leptin stimulation in TOLF cells but not in non-TOLF cells (Fig. 2, A and B). In addition, mineralization assays showed that whereas in the absence of leptin treatment, both TOLF cells and non-TOLF cells exhibited a fibroblast-like, spindle-shaped appearance and none of these cells manifested mineralization, under leptin stimulation the cell matrix began to mineralize, and crystals appeared at 4 weeks in 8 of 10 TOLF cell cultures, whereas no mineralization was observed in all 10 non-TOLF cell cultures (Fig. 2C). Collectively, all these experiments indicated that whereas leptin had no appreciable effect on the proliferation of both TOLF cells and non-TOLF cells, it stimulated osteogenic differentiation of TOLF cells but not non-TOLF cells.FIGURE 2Effects of leptin on osteocalcin proteins expression and ALP activity in TOLF and non-TOLF cells. Cells were treated with 10 ng/ml leptin for different periods of time (0–7 days), and cellular proteins were extracted for osteocalcin protein detection (A) and ALP activity measurement (B). Each bar represents the mean ± S.D. from 8 samples, and the asterisks indicate statistical significance (p < 0.05). Controls were cells maintained under resting state (0 days). C, comparison of TOLF and non-TOLF cells for the ability to form mineralized nodules. Cells were incubated in DMEM supplemented with 10% FBS. After cultures reached confluence, cells were maintained in DMEM medium containing 10% FBS, 10 mm β-glycerophosphate, and 100 ng/ml leptin for the indicated periods of time (0–4 weeks) and then stained with alizarin red.View Large Image Figure ViewerDownload Hi-res image Download (PPT) Molecular Events Involved in Leptin-stimulated Osteogenesis in TOLF Cells—To gain insight into the mechanism underlying the osteogenic effect of leptin, we first examined the expression of leptin receptors in TOLF cells and non-TOLF cells. These cells were grown in normal media for 72 h and harvested for total protein and total RNA extraction. The expression of leptin receptor messengers and proteins was examined by RT-PCR and Western blotting, respectively. As shown in Fig. 3A, similar expression levels of the mRNA and the protein of leptin receptor were detected in TOLF cells as well as in non-TOLF cells. As stated before, leptin is believed to exert its biological function through binding to its receptors, which in turn transduce the signal through the activation of STAT3, ERK, JNK, and p38 pathways (20Hegyi K. Fulop K. Kovacs K. Toth S. Falus A. Cell Biol. Int. 2004; 28: 159-169Crossref PubMed Scopus (200) Google Scholar, 21Sweeney G. Cell .Signal. 2002; 14: 655-663Crossref PubMed Scopus (309) Google Scholar). Thus, we next examined the status of STAT3, ERK, JNK, and p38 phosphorylation in response to leptin treatment in TOLF cells. Cellular proteins were extracted from TOLF cells that were exposed to leptin for various periods of time and were then immunoblotted with antibodies against phospho-STAT3, phospho-ERK1/2, phospho-JNK, and phospho-p38. These experiments showed that, whereas the total expression levels of STAT3, ERK1/2, and JNK were unchanged over the time of leptin treatment, the phosphorylation of STAT3, ERK1/2, and JNK was stimulated by leptin (Fig. 3B). However, leptin treatment did not affect both the expression and the phosphorylation of p38 in TOLF cells, although leptin was capable of stimulating p38 phosphorylation in breast cancer cells MCF-7 and T47-D in our experimental system (Fig. 3B). Collectively, these experiments indicated the osteogenic effect of leptin in TOLF cells could be mediated via STAT3, ERK1/2, and/or JNK signaling pathways. STAT3 Signaling Pathway Is Critically Involved in Leptin-induced Osteogenic Differentiation of TOLF Cells—To investigate whether activation of STAT3, ERK1/2, and JNK signaling pathways was indeed linked to the osteogenic effect of leptin in TOLF cells, we used selective inhibitors to block STAT3, ERK1/2, or JNK phosphorylation, respectively, and detected the expression of osteocalcin, a marker for cellular osteogenic differentiation, by real-time RT-PCR under the treatment of leptin. As shown in Fig. 4A, treatment of TOLF cells with Janus tyrosine kinase/STAT3 phosphorylation-selective inhibitor AG490 selectively inhibited the phosphorylation of STAT3 protein. Analogously, treatment of TOLF cells with ERK1/2 phosphorylation-selective inhibitor PD98059 and JNK phosphorylation-selective inhibitor SP600125 selectively inhibited the phosphorylation of ERK1/2 and JNK, respectively. Measurement of osteocalcin expression indicated that blocking phosphorylation of ERK1/2 and JNK had only marginal effects on osteocalcin expression, whereas blocking STAT3 phosphorylation resulted in a significant reduction of leptin-induced osteocalcin expression (Fig. 4B), suggesting that STAT3 signaling pathway is the major mediator for the osteogenic effect of leptin in TOLF cells. Transactivation of Osteocalcin by STAT3 in TOLF Cells under Leptin Treatment—STAT3 is a cytoplasmic protein, which once phosphorylated is translocated into the nucleus where it regulates gene expression through interacting with cofactors (22Lo H.W. Hsu S.C. Ali-Seyed M. Gunduz M. Xia W. Wei Y. Bartholomeusz G. Shih J.Y. Hung M.C. Cancer Cell. 2005; 7: 575-589Abstract Full Text Full Text PDF PubMed Scopus (426) Google Scholar). Runx2 is a key transcriptional factor in cell osteogenic differentiation. Activated Runx2 transactivates the transcription of its target gene such as osteocalcin (23Zaidi S.K. Sullivan A.J. Medina R. Ito Y. van Wijnen A.J. Stein J.L. Lian J.B. Stein G.S. EMBO J. 2004; 23: 790-799Crossref PubMed Scopus (333) Google Scholar), which in turn promotes cellular osteogenic differentiation (24Paredes R. Arriagada G. Cruzat F. Villagra A. Olate J. Zaidi K. van Wijnen A. Lian J.B. Stein G.S. Stein J.L. Montecino M. Mol. Cell. Biol. 2004; 24: 8847-8861Crossref PubMed Scopus (125) Google Scholar). The observation that osteocalcin expression was increased by leptin treatment in TOLF cells (Fig. 1, C and E) suggests that leptin-activated STAT3 could be functionally linked to Runx2 in these cells. To investigate a functional interaction between STAT3 and Runx2 in transactivating Runx2 target genes, thus in mediating the osteogenic effect of leptin, we first examined if STAT3 could interact with Runx2 in the nucleus of TOLF cells under leptin stimulation. Co-immunoprecipitation experiments were performed with nuclear extractions of TOLF cells treated with leptin using STAT3 antibodies, and the immunoprecipitates were then immunoblotted with antibodies against Runx2. As shown in Fig. 5A, a physical interaction between STAT3 and Runx2 was detected in the nucleus of TOLF cells upon the stimulation of leptin treatment, providing a basis for a functional interaction between STAT3 and Runx2. STAT3 activates gene transcription through recruitment of coactivators that modify the chromatin architecture (25Ying Q.L. Nichols J. Chambers I. Smith A. Cell. 2003; 115: 281-292Abstract Full Text Full Text PDF PubMed Scopus (1748) Google Scholar, 26Yoo J.Y. Huso D.L. Nathans D. Desiderio S. Cell. 2002; 108: 331-344Abstract Full Text Full Text PDF PubMed Scopus (115) Google Scholar, 27Bromberg J.F. Wrzeszczynska M.H. Devgan G. Zhao Y. Pestell R.G. Albanese C. Darnell Jr., J.E. Cell. 1999; 98: 295-303Abstract Full Text Full Text PDF PubMed Scopus (2549) Google Scholar, 28Wen Z. Zhong Z. Darnell Jr., J.E. Cell. 1995; 82: 241-250Abstract Full Text PDF PubMed Scopus (1770) Google Scholar). Previous studies have shown that the p160 family coactivators, SRC-1, GRIP1, and AIB1, although originally identified as cofactors involved in nuclear receptor-mediated gene transcription, also participate in gene transcriptional activation mediated by other transcription factors, including STATs (29Arimura A. vn Peer M. Schroder A.J. Rothman P.B. J. Biol. Chem. 2004; 279: 31105-31112Abstract Full Text Full Text PDF PubMed Scopus (49) Google Scholar, 30Goenka S. Marlar C. Schindler U. Boothby M. J. Biol. Chem. 2003; 278: 50362-50370Abstract Full Text Full Text PDF PubMed Scopus (16) Google Scholar, 31Oki S. Limnander A. Danial N.N. Rothman P.B. Blood. 2002; 100: 966-973Crossref PubMed Scopus (16) Google Scholar, 32Litterst C.M. Pfitzner E. J. Biol. Chem. 2002; 277: 36052-36060Abstract Full Text Full Text PDF PubMed Scopus (49) Google Scholar, 33Litterst C.M. Pfitzner E. J. Biol. Chem. 2001; 276: 45713-45721Abstract Full Text Full Text PDF PubMed Scopus (57) Google Scholar). To investigate whether the p160 family coactivators are also implicated in the osteogenic activity of leptin, transient transfection experiments were performed to examine the enhancement of Runx2 target gene transcription by p160 coactivators. In these experiments osteocalcin gene promoter-driven luciferase (pOC-LUC) (13Sierra J. Villagra A. Paredes R. Cruzat F. Gutierrez S. Javed A. Arriagada G. Olate J. Imschenetzky M. Van Wijnen A.J. Lian J.B. Stein G.S. Stein J.L. Montecino M. Mol. Cell. Biol. 2003; 23: 3339-3351Crossref PubMed Scopus (189) Google Scholar) was cotransfected with mammalian expression vectors for p160 coactivators SRC-1, GRIP1, or AIB1 in TOLF cells, and the luciferase activity was measured in the presence or absence of leptin stimulation. As shown in Fig. 5B, the experiments indicated that SRC-1, but not GRIP1 or AIB1, was able to enhance the expression of the luciferase gene. This discrepancy was not a result of the coactivator protein abundance because the expression levels of SRC-1, GRIP1, and AIB1 in these cells were similar (Fig. 5C) and the enhanced expression of endogenous osteocalcin gene by SRC-1 was also evident as measured by Western blot analysis (Fig. 5D). These observations suggested that there was a differential involvement of p160 coactivators in transcription of genes that are important in osteogenesis. Transactivation of Osteocalcin Gene by STAT3, Runx2, and SRC-1—To further dissect the leptin-activated STAT3 interaction with Runx2 and to understand the involvement of SRC-1 coactivator in Runx2 target gene coactivation, we examined the recruitment of STAT3, Runx2, and SRC-1 on the promoter of osteocalcin gene using ChIP assays (14Shang Y. Hu X. DiRenzo J. Lazar M.A. Brown M. Cell. 2000; 103: 843-852Abstract Full Text Full Text PDF PubMed Scopus (1469) Google Scholar). In these experiments soluble chromatins were prepared using a formaldehyde cross-linking protocol, and the occupancy of Runx2 target gene promoter by SRC-1, STAT3, and Runx2 was analyzed using specific antibodies against SRC-1, STAT3, or Runx2 and using a pair of primers spanning the Runx2-responsive region in the osteocalcin promoter (Fig. 5E, upper panel). Measurement of GRIP1 and AIB1 also was included in the experiment. As shown in Fig. 5E (lower panel), ChIP experiments detected the occupancy of SRC-1, but not GRIP1 or AIB1, along with STAT3 and Runx2 on the promoter of osteocalcin gene under the treatment of leptin. The increased presence of SRC-1 on osteocalcin promoter was not resulted from leptin-induced accumulation of SRC-1 in the cell nucleus as Western blotting did not detect any increase of nuclear SRC-1 with leptin treatment (Fig. 5F). We next performed Re-ChIP of the cross-linked and sonicated chromatin from TOLF cells with antibodies against STAT3 and then with anti-SRC-1, antibodies against SRC-1, and then with anti-STAT3 or antibodies against Runx2 and then with anti-SRC-1. This was followed by quantitative PCR analyses for determining whether osteocalcin promoter sequences were detected in the final precipitate (Fig. 5G). When chromatins were first immunoprecipitated with anti-STAT3 (Fig. 5G, column 3) and the collected samples were subsequently re-precipitated with anti-SRC-1 antibodies (Fig. 5G, column 7), the presence of osteocalcin promoter was detected in the precipitate. Similarly, chromatins first immunoprecipitated with anti-SRC-1 (Fig. 5G, column 4) and then re-precipitated with anti-STAT3 antibodies (Fig. 5G, column 8) revealed the presence of osteocalcin promoter. These results indicated that both STAT3 and SRC-1 were recruited to the promoter of osteocalcin gene. In addition, when chromatin extracts were sequentially immunoprecipitated with anti-Runx2 and then anti-SRC-1 antibodies (Fig. 5G, column 6), the osteocalcin promoter was also detected in the collected materials. Collectively, these results suggested that all STAT3, Runx2, and SRC-1 occupied the promoter and, thus, participated in transactivation of osteocalcin gene. The binding of Runx2 and STAT3 on osteocalcin promoter is also supported by electrophoretic mobility shift assays (Fig. 6A). Because SRC-1 was the only p160 coactivator involved in the leptin-induced osteocalcin activation, we sought to further substantiate the role of SRC-1 in leptin-induced osteogenesis of TOLF cells. We used RNA-mediated interference (RNAi) approach to silence the expression of SRC-1, and the effect of RNAi on the stimulation of osteocalcin transcription by leptin in TOLF cells was measured by real-time PCR. As shown in Fig. 6B, the expression of SRC-1 could be effectively silenced by a vector carrying specific sequence against the mRNA of SRC-1, and leptin-stimulated osteocalcin transcription was severely affected with silenced SRC-1 expression in TOLF cells. These results further supported that SRC-1 plays an important role in leptin signaling pathway that leads to the osteogenesis of TOLF cells. It is believed that obesity is a risk factor for TOLF. Although other endocrine abnormalities such as hyperinsulinemia, a feature of obese people, might account for TOLF, a recent study shows that hyperleptinemia is closely correlated with TOLF (5Shirakura Y. Sugiyama T. Tanaka H. Taguchi T. Kawai S. Biochem. Biophys. Res. Commun. 2000; 267: 752-755Crossref PubMed Scopus (26) Google Scholar, 34Epstein N. J. Spinal Disord. 1996; 9: 446-450PubMed Google Scholar, 35Miyamoto S. Yonenobu K. Ono K. Spine. 1993; 18: 2267-2270Crossref PubMed Scopus (31) Google Scholar), suggesting that leptin, an adipocyte-derived cytokine, is critical in connecting at molecular levels the phenotypical manifestation of obesity and the pathological development of TOLF. In supporting this notion, leptin has been reported to stimulate the osteogenic differentiation of various cell types (36Parhami F. Tintut Y. Ballard A. Fogelman A.M. Demer L.L. Circ. Res. 2001; 88: 954-960Crossref PubMed Scopus (305) Google Scholar, 37Thomas T. Gori F. Khosla S. Jensen M.D. Burguera B. Riggs B.L. Endocrinology. 1999; 140: 1630-1638Crossref PubMed Google Scholar, 38Gordeladze J.O. Drevon C.A. Syversen U. Reseland J.E. J. Cell. Biochem. 2002; 85: 825-836Crossref PubMed Scopus (359) Google Scholar). We reported here the molecular events involved in leptin-stimulated osteogenesis in TOLF cells. We showed that leptin treatment induced the expression of osteocalcin and ALP as well as in vitro mineralization in TOLF cell cultures. In addition, we demonstrated that the STAT3 signaling pathway is activated in response to leptin treatment in TOLF cells and STAT3, along with steroid receptor coactivator SRC-1 and osteogenic transcription factor Runx2, transactivates osteocalcin, one of the Runx2 downstream target genes that may in turn mediate the osteogenic activity of leptin. STAT3 plays an essential role during embryonic development, cell survival, and differentiation (39Lee Y.J. Park J.H. Ju S.K. You K.H. Ko J.S. Kim H.M. FEBS Lett. 2002; 528: 43-47Crossref PubMed Scopus (66) Google Scholar). At the molecular level, STAT3 acts as a transcription activator and interacts with other nuclear factors to regulate a number of genes that are critically involved in cell proliferation and differentiation (40Theilhaber J. Connolly T. Roman-Roman S. Bushnell S. Jackson A. Call K. Garcia T. Baron R. Genome Res. 2002; 12: 165-176Crossref PubMed Scopus (65) Google Scholar, 41O'Brien C.A. Gubrij I. Lin S.C. Saylors R.L. Manolagas S.C. J. Biol. Chem. 1999; 274: 19301-19308Abstract Full Text Full Text PDF PubMed Scopus (260) Google Scholar). The biological function of STAT3 in osteotropic activity has been well documented. The gp130 knock-in mice gp130F759/F759, in which interleukin-6 family cytokine-mediated STAT3 activation is enhanced, showed an osteosclerotic phenotype, whereas mice with osteoblast-specific disruption of the STAT3 gene exhibited an osteoporotic phenotype (42Itoh S. Udagawa N. Takahashi N. Yoshitake F. Narita H. Ebisu S. Ishihara K. Bone (NY). 2006; 39: 505-512Crossref PubMed Scopus (138) Google Scholar). Runx2, also referred to as Cbfa1, Osf2, AML3, and PEBP2αA, is an osteoblast-related transcription factor that is essential for bone formation (43Salingcarnboriboon R. Tsuji K. Komori T. Nakashima K. Ezura Y. Noda M. Endocrinology. 2006; 147: 2296-2305Crossref PubMed Scopus (41) Google Scholar). Homozygous deletion of Runx2 arrests osteoblast maturation, resulting in the absence of endochondral and intramembranous ossification (44Dobreva G. Chahrour M. Dautzenberg M. Chirivella L. Kanzler B. Farinas I. Karsenty G. Grosschedl R. Cell. 2006; 125: 971-986Abstract Full Text Full Text PDF PubMed Scopus (431) Google Scholar). Runx2 can directly stimulate osteogenic differentiation by binding to an osteoblast-specific cis-acting element, termed OSE2, in the promoter region of skeletal target genes and regulating their expression (45Pockwinse S.M. Rajgopal A. Young D.W. Mujeeb K.A. Nickerson J. Javed A. Redick S. Lian J.B. van Wijnen A.J. Stein J.L. Stein G.S. Doxsey S.J. J. Cell. Physiol. 2006; 206: 354-362Crossref PubMed Scopus (52) Google Scholar). Our experiments identifying a physical and functional interaction between STAT3 and Runx2 in TOLF cells add another line of evidence to support the important roles of STAT3 and Runx2 in osteogenesis. The p160 coactivator family is thought to contribute to transcriptional activation by being recruited to the gene promoter and forming a platform for the binding of other coactivators. Initially identified as coactivators for nuclear receptor-mediated gene transcription, recent studies have shown that the p160 coactivators also are involved in transcriptional activation by various other transcription factors, such as activator protein, serum response factor, NFκB, and STATs (46Shang Y. Brown M. Science. 2002; 295: 2465-2468Crossref PubMed Scopus (1008) Google Scholar, 47Shang Y. Nat. Rev. Cancer. 2006; 6: 360-368Crossref PubMed Scopus (210) Google Scholar), implying a functional complexity for these proteins. We showed that one member of the p160 family, SRC-1, but not GRIP1 and AIB1, was recruited to Runx2 target gene promoter and coactivated the gene transcription, indicating that SRC-1 plays an important role in leptin signaling pathway that leads to the osteogenesis in TOLF cells. Leptin receptors are expressed in both TOLF cells and non-TOLF cells. Nonetheless, the osteogenic effect of leptin is different in TOLF cells versus non-TOLF cells. Moreover, even different TOLF cells exhibited different levels of mineralization in response to leptin treatment in our experiments. It is conceivable that osteogenic commitment of the cells in the ossification process of TOLF and the metaplasia of TOLF cells into osteogenic cells have already occurred in TOLF, which is consistent with other studies of OLF pathogenesis (19Nakamura T. Hashimoto N. Maeda Y. Ikeda T. Nakagawa H. Takagi K. J. Spinal Disord. 1990; 3: 288-292Crossref PubMed Google Scholar, 48Akhaddar A. Mansouri A. Zrara I. Gazzaz M. Maftah M. Mostarchid B. Benomar S. Boucetta M. Joint Bone Spine. 2002; 69: 319-323Crossref PubMed Scopus (19) Google Scholar). Further studies are warranted to investigate the molecular mechanism underlying osteogenic commitment of TOLF cells and molecular events involved in the initiation of TOLF.
What problem does this paper attempt to address?
-
Surgical treatment of 40 patients with thoracic ossification of the ligamentum flavum.
Fangcai Li,Qixin Chen,Kan Xu
DOI: https://doi.org/10.3171/spi.2006.4.3.191
2007-01-01
Abstract:Object. The authors evaluated different surgical methods used to treat thoracic ossification of the ligamentum flavum (OLF). Methods. Data obtained in 40 patients who underwent posterior decompression for thoracic myelopathy caused by thoracic OLF were studied retrospectively. There were 32 men and eight women. All patients underwent posterior decompression in which laminoplasty was performed or laminectomy combined with lateral fusion. Every surgical specimen was stained with H & E, and scanning electron microscopy was performed in 20 cases. The mean follow-up period was 28 months. Postoperative outcomes were evaluated using a recovery scale based on the Japanese Orthopaedic Association classification. There were a total of 168 ossified segments in this series, 77.4% of which were located in the lower thoracic spine. Marginal osteophyte formation was found in 36 patients; in 32 of the 36 patients, these marginal osteophytes were located at the intervertebral space either higher or lower than the ossified segment. Scanning electron microscopy showed elastic fiber breakdown, proliferation of collagenous fibers, calcification, and OLF in the same microscopy region. Laminoplasty was performed in four patients. In three cases surgery resulted in unchanged or worse outcome (increased kyphotic deformity in two), and in one it resulted in good outcome. Laminectomy combined with lateral fusion was performed in 36 patients, in 30 cases of which it resulted in a good or fair outcome, and increased kyphotic deformity in only one. Of these 36 laminectomy-treated patients, an en bloc laminectomy-treated procedure was performed in 16 patients; in 11 of the 12 patients with lateral or diffuse-type lesions the surgery resulted in a good or fair outcome. En bloc laminectomy, however, seems ineffective in the treatment of patients with thickened, nodular-type thoracic OLF, as the procedure resulted in worse outcome in two of the four patients. The authors have thus developed a new modality of laminectomy that they have termed "separating laminectomy," which they performed in 16 patients with thickened, nodular-type OLF; in 13 cases it resulted in a good or fair outcome, and in only one case did it result in a worse outcome. Conclusions. The pathogenesis of thoracic OLF is mainly due to the localized mechanical stress on the ligament. Laminectomy combined with lateral fusion is the treatment of choice for thoracic OLF; furthermore, in terms of the configuration of the ossified lesion, en bloc laminectomy is suitable for the treatment of lateral-type and diffuse-type OLF, and the separating laminectomy is suitable for the thickened, nodular-type OLF.
-
Combined use of leptin and mechanical stress has osteogenic effects on ossification of the posterior longitudinal ligament
Shuai Chen,Haifeng Zhu,Gangliang Wang,Ziang Xie,Jiying Wang,Jian Chen
DOI: https://doi.org/10.1007/s00586-018-5663-4
2018-01-01
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. Graphical abstract These slides can be retrieved under Electronic Supplementary Material.
-
Fibrotic and Inflammatory Characteristics of Epidural Fat Adjacent to the Ossification Area in Patients with Ossification of the Ligament Flavum
Xinyu Dou,Tianli Mao,Yunlong Ma,Fei Jia,Yu Liu,Xiaoguang Liu
DOI: https://doi.org/10.1002/jsp2.1229
2022-01-01
JOR Spine
Abstract:AbstractObjectivesTo observe histological and inflammatory characteristics of epidural fat (EF) adjacent to the ossification area in patients with ossification of the thoracic ligament flavum (TOLF) and provide a preliminary research basis for investigating the impact of the EF on OLF.MethodsSamples of EF and autologous subcutaneous adipose tissue (SCAT) from TOLF patients (n = 26) and non‐TOLF patients (n = 23) were harvested during posterior thoracic spine surgery. Adipocyte size and fibrosis were measured by histology. Vascularization and inflammatory cell infiltration were evaluated by immunohistochemical staining. Adipogenesis‐related genes were assessed by real‐time quantitative PCR. Conditioned media from cultured EF were evaluated via enzyme‐linked immunosorbent assay to detect the secretion of inflammatory cytokines, including interleukin‐6 (IL‐6), tumor necrosis factor (TNF‐α), and leptin. The phosphorylated STAT3 protein level in ligament flavum (LF) was examined using Western blot.ResultsAdipocytes size in EF was similar between in the TOLF and non‐TOLF groups, but significantly smaller than that from autologous SCAT. Adipogenesis‐related mRNA expression in EF was lower than that in SCAT. More fibrosis and vascularization were found in TOLF than in non‐TOLF. EF in the TOLF group exhibited more macrophages and B lymphocytes infiltrated. The levels of cytokines such as IL‐6, TNF‐α, and leptin secreted by EF were significantly higher in the TOLF group than non‐TOLF. The level of phosphorylated STAT3 in LF was significantly upregulated in the TOLF group.ConclusionsMorphologically, EF adjacent to the ossification area is smaller and more uniform than autologous SCAT, exhibiting a characteristic similarity to visceral fat. EF in the TOLF group shows a more fibrotic, vascularized, and inflammatory phenotype, which secretes multiple cytokines. The phosphorylated STAT3 protein was significantly upregulated in the TOLF group. Whether these properties of EF directly affect the process of OLF needs to be further explored.
-
The Involvement And Possible Mechanism Of Pro-Inflammatory Tumor Necrosis Factor Alpha (Tnf-Alpha) In Thoracic Ossification Of The Ligamentum Flavum
Chi Zhang,Zhongqiang Chen,Xiangyu Meng,Mengtao Li,Li Zhang,Ann Huang
DOI: https://doi.org/10.1371/journal.pone.0178986
IF: 3.7
2017-01-01
PLoS ONE
Abstract:Thoracic ossification of the ligamentum flavum (TOLF) is characterized by ectopic bone formation in the ligamentum flavum and is considered to be a leading cause of thoracic spinal canal stenosis and myelopathy. However, the underlying etiology is not well understood. An iTRAQ proteomics was used to reveal the involvement of inflammation factors in TOLF. TNF-alpha is a pro-inflammatory cytokine implicated in the pathogenesis of many human diseases. Protein profiling analysis showed that the protein level of TNF-alpha increased in the ossified ligamentum flavum of TOLF, which was confirmed by western blot. The effects of TNF-alpha on primary ligamentum flavum cells was examined. Cell proliferation assay demonstrated that primary cells from the ossified ligamentum flavum of TOLF grew faster than the control. Flow cytometry assay indicated that the proportions of cells in S phase of cell cycle of primary cells increased after TNF-alpha stimulation. To address the effect of TNF-alpha on gene expression, primary cells were derived from ligamentum flavum of TOLF patients. Culture cells were stimulated by TNF-alpha. RNA was isolated and analyzed by quantitative RT-PCR. G1/S-specific proteins cyclin D1 and c-Myc were upregulated after TNF-alpha stimulation. On the other hand, osteoblast differentiation related genes such as Bmp2 and Osterix (Osx) were upregulated in the presence of TNF-alpha. TNF-alpha activated Osx expression in a dose dependent manner. Interestingly, a specific mitogen-activated protein kinase ERK inhibitor U0126, but not JNK kinase inhibitor SP600125, abrogated TNF-alpha activation of Osx expression. This suggests that TNF-alpha activates Osx expression through the mitogen-activated protein kinase ERK pathway. Taken together, we provide the evidence to support that TNF-alpha involves in TOLF probably through regulating cell proliferation via cyclin D1 and c-Myc, and promoting osteoblast differentiation via Osx.
-
Cyclic Tensile Stress to Rat Thoracolumbar Ligamentum Flavum Inducing the Ossification of Ligamentum Flavum
Yin Zhao,Bo Yuan,Li Cheng,Shengyuan Zhou,Yifan Tang,Zhichao Zhang,Yanqing Sun,Zheng Xu,Fengning Li,Xinyuan Liao,Xiongsheng Chen
DOI: https://doi.org/10.1097/brs.0000000000004087
IF: 3.241
2021-05-04
Spine
Abstract:STUDY DESIGN: Western blot, reverse transcription-polymerase chain reaction (RT-PCR), radiological, and histological analyses of the rat ossification of ligamentum flavum (OLF) induced by cyclic tensile stress.OBJECTIVE: The aim of this study was to induce the OLF using cyclic tensile stress to rat thoracolumbar ligamentum flavum, and to investigate the possible molecular mechanism of tension-induced OLF.SUMMARY OF BACKGROUND DATA: Tensile stress has been considered as an important factor leading to the OLF. So far, however, no OLF induced by tension has been reported.METHODS: Forty rats were randomly divided into five equal groups. For control groups, the blank and anesthesia groups were not subjected to tension. For experimental groups, the 4-, 8-, and 12-week groups were subjected to cyclic tensile stress of ligamentum flavum after abdominal anesthesia for 4 weeks, 8 weeks, and 12 weeks, respectively, using an original stress apparatus for rats. The radiological and morphological changes of rat spine, as well as the protein and mRNA expressions of CD44, bone morphogenetic protein-2 (BMP-2), integrin β3, collagen protein type I (COL1), osteopontin (OPN), runt-related transcription factor 2 (RUNX-2), and vascular endothelial growth factor (VEGF), were concerned.RESULTS: The micro-CT showed OLF in the 4-, 8-, and 12-week group. The axial maximum occupied area of ossifications was 1.42 mm2, 3.35 mm2, and 7.28 mm2, respectively. In histopathology, chondrocytes proliferated in the experimental model; woven bone arose in the 8- and 12-week groups, and was more noticeable in the 12-week group. According to western blot and RT-PCR, the expressions of seven osteogenesis-related molecules were all increased in three experimental groups.CONCLUSION: Cyclic tensile stress to the ligamentum flavum in rats can induce the OLF, and the longer the duration, the more visible the osteogenesis. The upregulation and synergism of osteogenesis-related molecules may contribute to the OLF induced by tensile stress.Level of Evidence: N/A.
clinical neurology,orthopedics
-
Itraq Quantitative Proteomic Study in Patients with Thoracic Ossification of the Ligamentum Flavum.
Bingxiang Wang,Zhongqiang Chen,Xiangyu Meng,Mengtao Li,Xiaoxi Yang,Chi Zhang
DOI: https://doi.org/10.1016/j.bbrc.2017.04.136
IF: 3.1
2017-01-01
Biochemical and Biophysical Research Communications
Abstract:Thoracic ossification of the ligamentum flavum (TOLF) is a unique disease with ectopic ossification, and is a major cause of thoracic spinal stenosis and myelopathy. However, the underlying etiology remains largely unknown. In this study, the ligamentum flavum was systematically analyzed in TOLF patients by using comprehensive iTRAQ labeled quantitative proteomics. Among 1285 detected proteins, there were 282 proteins identified to be differentially expressed. The Gene Ontology (GO) analysis regarding functional annotation of proteins consists of the following three aspects: the biological process, the molecular function, and the cellular components. The function clustering analysis revealed that ten of the above proteins are related to inflammation, such as tumor necrosis factor (TNF). This finding was subsequently validated by ELISA, which indicated that serum TNF-α of TOLF patients was significantly higher compared with the control group. To address the effect of TNF-α on ossification-related gene expression, we purified and cultured primary cells from thoracic ligamentum flavum of patients with TOLF. TNF-α was then used to stimulate cells. RNA was isolated and analyzed by RT-PCR. Our results showed that TNF-α was able to induce the expressions of osteoblast-specific transcription factor Osterix (Osx) in ligamentum flavum cells, suggesting that it can promote osteoblast differentiation. In addition, as the Osx downstream osteoblast genes OCN and ALP were also activated by TNF-α. This is the first proteomic study to identify inflammation factors such as TNF-α involved in ossified ligamentum flavum in TOLF, which may contribute to a better understanding of the cause of TOLF.
-
Interleukin-17A Promotes Proliferation and Osteogenic Differentiation of Human Ligamentum Flavum Cells Through Regulation of Β-Catenin Signaling.
Jialiang Lin,Shuai Jiang,Qian Xiang,Yongzhao Zhao,Longjie Wang,Dongwei Fan,Woquan Zhong,Chuiguo Sun,Zhongqiang Chen,Weishi Li
DOI: https://doi.org/10.1097/brs.0000000000004789
2023-01-01
Abstract:Study Design. A basic experimental study.Objective. To elucidate the role and mechanism of interleukin (IL)-17A in thoracic ossification of the ligamentum flavum (TOLF).Summary of Background Data. TOLF is characterized by the replacement of the thoracic ligamentum flavum with ossified tissue and is one of the leading causes of thoracic spinal stenosis. IL-17A is an important member of the IL-17 family that has received widespread attention for its key contributions to the regulation of bone metabolism and heterotopic ossification. However, it is unclear whether IL-17A is involved in TOLF.Materials and Methods. Cell counting kit-8 assay and 5-ethynyl-2'-deoxyuridine staining were performed to assess the proliferation of ligamentum flavum cells (LFCs). Alkaline phosphatase activity assay, Alizarin red staining, and protein level expression of osteogenic-related genes were used to evaluate the osteogenic differentiation potential of LFCs. The effect of IL-17A on the proliferation and osteogenic differentiation of LFCs was further assessed after silencing beta-catenin by transfection with small interfering RNA. In addition, the possible source of IL-17A was further demonstrated by coculture assays of T helper 17 (Th17) cells with LFCs. Student t test was used for comparisons between groups, and the one-way analysis of variance, followed by the Tukey post hoc test, was used for comparison of more than two groups.Results. IL-17A was elevated in TOLF tissue compared with normal ligamentum flavum. IL-17A stimulation promoted the proliferation and osteogenic differentiation of LFCs derived from patients with TOLF. We found that IL-17A promoted the proliferation and osteogenic differentiation of LFCs by regulating the beta-catenin signaling. Coculture of Th17 cells with LFCs enhanced beta-catenin signaling-mediated proliferation and osteogenic differentiation of LFCs. However, these effects were markedly attenuated after the neutralization of IL-17A.Conclusions. This is the first work we are aware of to highlight the importance of IL-17A in TOLF. IL-17A secreted by Th17 cells in the ligamentum flavum may be involved in the ossification of the microenvironment by regulating beta-catenin signaling to promote the proliferation and osteogenic differentiation of LFCs.
-
IL-6 is Involved in Thoracic Ossification of the Ligamentum Flavum
Ann Yehong Huang,Li Shu,Zhongqiang Chen,Chi Zhang
DOI: https://doi.org/10.1371/journal.pone.0272357
IF: 3.7
2022-01-01
PLoS ONE
Abstract:Thoracic ossification of the ligamentum flavum (TOLF) is a heterotopic ossification of spinal ligaments. TOLF is the major cause of thoracic spinal canal stenosis and myelopathy, and its underlying mechanisms are not clear. Bone formation is a complex developmental process involving the differentiation of mesenchymal stem cells to osteoblasts, and regulated by BMP2, RUNX2, Osterix (OSX), etc. In this study, we continue to further characterize properties of TOLF. Our immunohistochemistry experiments showed that expressions of osteoblastic factors such as BMP2 and RUNX2 increased in TOLF. According to flow cytometry analysis the proportion of S phase of cell cycle in primary TOLF cells was 9% higher than the control. Alizarin red staining and ALP staining observations were consistent with immunohistochemistry results. It was also observed that inflammatory cytokine IL-6 level dramatically increased in the culture supernatant of primary TOLF cells. We propose the hypothesis that IL-6 is involved in TOLF. To testify the hypothesis, we examined the effect of IL-6. Our results showed that IL-6 was able to activate expressions of osteoblastic factors such as BMP2, RUNX2, OSX, OCN and ALP, and that expressions of cell proliferation factors cyclin D1 and cyclin C increased in the presence of IL-6. Moreover, IL-6-induced BMP2 expression was inhibited by p38 inhibitor SB203580, indicating that IL-6 regulated the osteogenic BMP2 activation through p38 MAPK pathway. These data suggest that IL-6 is involved in TOLF.
-
The evaluation of osteogenic potency of ligament cells in thoracic ossification of the ligamentum flavum and relevant transcriptome high-throughput sequencing analysis
Shanglong Ning,Zhongqiang Chen,Xinlong Ma,Dongwei Fan,Chuiguo Sun,Jun Miao,Yan Zeng,Weishi Li
DOI: https://doi.org/10.3760/cma.j.issn.0253-2352.2017.20.008
2017-01-01
Abstract:Objective To investigate the osteogenic differentiation potency of ligament cells in thoracic ossification of the ligamentum flavum (TOLF) and analyze further by using transcriptome high-throughput sequencing.Methods Clinically,the patients with non-TOLF and TOLF (n=10 in each group) who underwent surgery in our hospital from October 2015 to April 2016 were included in this study.The primary ligament cells that derived from the two groups were separately cultured and induced osteogenesis with 15% strength of cyclic mechanical stress for 12h and 24h using a device called Flexcell FX-4000.The ALP activity was determined to evaluate the osteogenesis using quantitative analysis and ALP staining assay.Real-time PCR and westernblotting were used to detect the mRNA and protein expression of osteogenic-related genes including ALP,BMP-2 and Osteocalcin.Then,three patients in each group were included in the study of transcriptome high-throughput sequencing and bioinformatics analysis using Illumina HiSeqTM 2500 sequencing platform to compare further.Results The morphology of the cells that derived from two groups was basically similar,all presented an elongate spindle-shape.To evaluate the ostogenesis,ALP activity assays including quantitative and staining assays were performed.Under microscope,the ALP staining in the TOLF group was higher than non-TOLF group and increased with the longer duration of stress induction.The result of semi-quantitative analysis showed the stained area and positive cells in TOLF group were more than non-TOLF group significantly at 0 h,and were increased with the induction.The results of quantitative analysis showed ALP activity in the TOLF group was significantly higher than non-TOLF group and were increased with the induction significantly all the time.But no significant change in ALP staining or quantitative analysis was found in non-TOLF.The results of real-time PCR indicated that the expression of ostegenic markers above in the TOLF group was more than non-TOLF group significantly except the expression of OCN at 0 h.The expression of the three ostegenic markers in TOLF group was increased with the stress induction for 12 h and 24 h significantly except the expression of BMP-2 and OCN at 12 h.The results of western-blotting indicated that the expression of the three ostegenic markers above in the TOLF group was more than non-TOLF group significantly except the expression of ALP at 0 h.The expression of the three ostegenic markers in TOLF group was increased with the stress induction for 12 h and 24 h,but only the expression of ALP at 24 h was significant.And no significant change in the expression of mRNA and protein was found in non-TOLF group.In the transcriptome analysis,671 genes of TOLF group were up-regulated and 314 genes were found to be down-regulated compared to the control group.In addition,22 significant GO terms associated with upregulated genes were found to be closely related to ossification.Conclusion TOLF ligament cells have high osteogenic differentiation potency,which could express obvious osteogenesis-related gene spectrum,and differentially expressed genes including L1RL1 、PTHLH、DKK1 、BMP6、SPP1 and FGF1 may be related with the osteogenic potency of ligament cells in thoracic ossification.
-
Research progress of molecular signaling pathways in thoracic ossification of ligamentum flavum
张保良,陈仲强
DOI: https://doi.org/10.3760/cma.j.cn121113-20200222-00091
2020-01-01
Abstract:Thoracic ossification of ligament flavum (TOLF) is a chronic progressive disease presenting the heterotopic ossification of the ligamentum flavum of the thoracic spine, which leads to a series of symptoms and signs of thoracic spinal cord, nerveor vascular compression, with a high incidence in East Asian countries such as China and Japan. The pathology of TOLF is endochondral ossification in nature and surgical intervention is currently the only effective treatment due to conservative treatment does not work. The older age, the insidious onset, the various clinical manifestations of TOLF patients make the localization diagnosis and surgery management difficult and even bring about the high incidence of postoperative complications. Therefore, it is particularly important to study the mechanism of osteogenic differentiation of TOLF to find a way of early diagnosis and prevention. A variety of factors related to osteogenic differentiation of TOLF have been found, such as genetics, mechanical stimuli, molecular biological factors, metabolic and trace element abnormalities and so on. Moreover, molecular signaling pathways play a crucial role in regulating cell proliferation and directed differentiation. Therefore, this article reviews the confirmed relevant signaling pathways in the process of ligamentum flavum ossification, including bone morphogenetic protein (BMP) signaling pathway, mitogen-activated protein kinase (MAPK) signaling pathway, Wnt signaling pathway, Notch signaling pathway, STAT3 signaling pathway and other signaling pathways and factors, and also summarizes the promotion and inhibition of these signaling pathways in the osteogenic differentiation of ligamentum flavum cells, aiming at establishing a basis for the further research of the molecular biological mechanism of TOLF, and providing a possible direction for exploration of effective preventive and therapeutic targets.
-
Pathogenesis of Thoracic Ossification of the Ligamentum Flavum
Chi Zhang,Yanan Chang,Li Shu,Zhongqiang Chen
DOI: https://doi.org/10.3389/fphar.2024.1496297
IF: 5.6
2024-01-01
Frontiers in Pharmacology
Abstract:Thoracic ossification of the ligamentum flavum (TOLF) is characterized by ectopic ossification of the ligamentum flavum in the thoracic spine and is considered the main cause of thoracic spinal stenosis and spinal cord disease. Osteoblast specific transcription factor Osterix (Osx) is required for bone formation, and there is no bone formation or ossification without Osx. Surgical intervention is recognized as the only effective method for TOLF treatment with set of complications. However, underlying mechanisms of TOLF are not well understood. This paper summarizes the pathogenesis of TOLF. Some relevant factors have been discussed, such as mechanical stress, genetic susceptibility genes, endocrine and trace element metabolism abnormalities, which may associate with TOLF. More recent studies using proteomics technology and RNA sequencing approach have discovered that some new factors participate in TOLF by upregulation of Osx gene expression including inflammatory factors. TOLF is a unique disease involving multiple factors. On the other hand, studies on TOLF pathogenic mechanism may provide new ideas for finding possible upstream regulatory factors of Osx and further developing novel drugs to stimulate new bone formation to treat osteoporosis.
-
Genome‐wide DNA Methylation Profile Analysis in Thoracic Ossification of the Ligamentum Flavum
Tianqi Fan,Xiangyu Meng,Chuiguo Sun,Xiaoxi Yang,Guanghui Chen,Weishi Li,Zhongqiang Chen
DOI: https://doi.org/10.1111/jcmm.15509
2020-01-01
Journal of Cellular and Molecular Medicine
Abstract:Thoracic ossification of the ligamentum flavum (TOLF) causes serious spinal canal stenosis. The underlying aetiology may relate to genetic and inflammatory factors. DNA methylation plays a critical role in osteogenesis and inflammation, whereas there is no genome‐wide DNA methylation analysis about TOLF. The two subtypes of TOLF (single‐level and multiple‐level) have distinct clinical features. Using micro‐computed tomography (micro‐CT), we showed the ossification arose from the joint between two vertebrae at one/both sides of ligament flavum. With Illumina Infinium Human Methylation 850 BeadChip arrays, genome‐wide DNA methylation profile was measured in ligament flavum of eight healthy and eight TOLF samples. Only 65 of the differentially methylated cytosine‐phosphate‐guanine dinucleotides were found in both subtype groups. Principal component analysis and heat map analysis showed a different methylation pattern in TOLF samples, and methylation patterns of two subtypes are also distinct. The Gene Ontology enrichment analysis was significantly enriched in differentiation and inflammation. Pyrosequencing analysis and quantitative real‐time polymerase chain reaction were performed to validate the arrays results and expression levels, to test six differentially methylated genes (SLC7A11, HOXA10, HOXA11AS, TNIK, homeobox transcript antisense RNA, IFITM1), using another independent samples (P < 0.05). Our findings first demonstrated an altered Genome‐wide DNA methylation profile in TOLF, and implied distinct methylated features in two subtypes.
-
Angiopoietin-2 Promotes Osteogenic Differentiation of Thoracic Ligamentum Flavum Cells Via Modulating the Notch Signaling Pathway
Xiaoxi Yang,Zhongqiang Chen,Xiangyu Meng,Chuiguo Sun,Mengtao Li,Li Shu,Dongwei Fan,Tianqi Fan,Ann Y. Huang,Chi Zhang
DOI: https://doi.org/10.1371/journal.pone.0209300
IF: 3.7
2018-01-01
PLoS ONE
Abstract:Thoracic ossification of the ligamentum flavum (TOLF) is heterotopic ossification of spinal ligaments, which may cause serious thoracic spinal canal stenosis and myelopathy. However, the underlying etiology remains inadequately understood. In this study, the ossification patterns of TOLF were analyzed by micro-computer tomography (micro-CT). The expression profile of genes associated with angiogenesis was analyzed in thoracic ligamentum flavum cells at sites of different patterns of ossification using RNA sequencing. Significant differences in the expression profile of several genes were identified from Gene Ontology (GO) analysis. Angiopoietin-2 (ANGPT2) was significantly up-regulated in primary thoracic ligamentum flavum cells during osteogenic differentiation. To address the effect of ANGPT2 on Notch signaling and osteogenesis, ANGPT2 stimulation increased the expression of Notch2 and osteogenic markers of primary thoracic ligamentum flavum cells of immature ossification, while inhibition of ANGPT2 exhibited opposite effect on Notch pathway and osteogenesis of cells of mature ossification. These findings provide the first evidence for positive regulation of ANGPT2 on osteogenic differentiation in human thoracic ligamentum flavum cells via modulating the Notch signaling pathway.
-
Interleukin-17A promotes proliferation and osteogenic differentiation of human ligamentum flavum cells through regulation of β-catenin nuclear translocation
Jialiang Lin,Shuai Jiang,Qian Xiang,Yongzhao Zhao,Longjie Wang,Dongwei Fan,Woquan Zhong,Chuiguo Sun,Zhongqiang Chen,Weishi Li
DOI: https://doi.org/10.21203/rs.3.rs-1874618/v1
2022-01-01
Abstract:Abstract Thoracic ossification of the ligamentum flavum (TOLF) is the leading cause of thoracic spinal stenosis with no specifc treatment available. IL-17A has received widespread attention for its key contribution in the regulation of bone metabolism and heterotopic ossification. However, it is unclear whether IL-17A is involved in TOLF. Our results showed that IL-17A expression was elevated in ossified ligamentum flavum tissue compared to normal ligamentum flavum. In in vitro experiments, IL-17A stimulation promoted the proliferation and osteogenic differentiation of ligamentum flavum cells (LFCs) derived from TOLF patients. Mechanistically, we found that IL-17A stimulation promoted nuclear translocation of β-catenin, whereas silencing of β-catenin attenuated IL-17A-induced proliferation and osteogenic differentiation of LFCs. Furthermore, co-culture of T helper 17 (Th17) cells with LFCs showed that IL-17A secreted by Th17 cells significantly enhanced the accumulation of β-catenin in the nucleus, as well as the proliferation and osteogenic differentiation of LFCs. However, these effects were markedly attenuated after the neutralization of IL-17A. Our study demonstrates that IL-17A secreted by Th17 cells in the lesion microenvironment may be involved in TOLF by regulating nuclear translocation of β-catenin to promote proliferation and osteogenic differentiation of LFCs. This study helps to elucidate the pathological mechanism of TOLF and suggests the possibility of IL-17A as a potential target for the prevention and treatment of TOLF.
-
Notch Signaling Pathways in Human Thoracic Ossification of the Ligamentum Flavum
Xiaochen Qu,Zhongqiang Chen,Dongwei Fan,Chuiguo Sun,Yan Zeng,Xiaofei Hou,Shanglong Ning
DOI: https://doi.org/10.1002/jor.23303
2016-01-01
Journal of Orthopaedic Research
Abstract:This study investigated the pathological process of Notch signaling in the osteogenesis of ligamentum flavum tissues and cells, and the associated regulatory mechanisms. Notch receptors, ligands, and target genes were identified by quantitative polymerase chain reaction (qPCR) in ligamentum flavum cells and immunohistochemistry in ligamentum flavum sections from ossification of the ligamentum flavum (OLF) patients and controls. The temporospatial expression patterns of JAG1/Notch2/HES1 in human ligamentum flavum cells during osteogenic differentiation were determined by qPCR. Lentiviral vectors for Notch2 overexpression and knockdown were constructed and transfected into ligamentum flavum cells before osteogenic differentiation to examine the function of Notch signaling pathways in the osteogenic differentiation of ligamentum flavum cells. Alkaline phosphatase, Runx2, Osterix, osteocalcin, and osteopontin mRNA levels, alkaline phosphatase activity, and Alizarin Red staining were used as indicators of osteogenic differentiation. JAG1/Notch2/HES1 mRNA levels were up‐regulated in ligamentum flavum cells from OLF patients, which increased during osteogenic differentiation. Immunohistochemical analysis suggested positive Notch2 expression at the ossification front. Down‐regulation of Notch2 expression decelerated osteogenic differentiation of ligamentum flavum cells, and Notch2 overexpression promoted osteogenic differentiation of ligamentum flavum cells. Expression of Runx2 and Osterix increased in a manner similar to that of Notch2 during osteogenic differentiation of ligamentum flavum cells, and Notch2 knockdown and overexpression influenced their expression levels. Notch signaling plays an important role in OLF, and Notch may affect the osteogenic differentiation of ligamentum flavum cells via interactions with Runx2 and Osterix.© 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1481–1491, 2016.
-
The involvement of epidural fat in ossification of the ligamentum flavum: From the perspective of exosomal proteome
Chao Wang,Yida Wang,Weihang Zhu,Qian Tang,Xuekang Wang,Lu Zhang
DOI: https://doi.org/10.1016/j.heliyon.2024.e34755
IF: 3.776
2024-07-22
Heliyon
Abstract:Ossification of the ligamentum flavum (OLF) is the primary etiology of thoracic spinal stenosis. The functional properties of epidural fat (EF), an adipose tissue located in close proximity to ligamentum flavum (LF), have been scarcely investigated. The metabolic state of adipocytes significantly influences their functionality, and exosomes play a pivotal role in intercellular communication. This study aimed to investigate the role of EF-derived exosomes in OLF and characterize their protein profile by proteomics analysis. Our findings demonstrate that exosomes obtained from EF adjacent to OLF possess the ability to enhance osteogenesis of fibroblasts in vitro. Furthermore, proteomics analysis revealed metabolic dysfunction in EF adipocytes and identified lactate dehydrogenase A (LDHA) as a potential mediator involved in the development of OLF. This study provides new insights into the pathogenic mechanism underlying OLF and offers a theoretical basis for preventing and treating ligament ossification.
-
Significance of Body Mass Index on Thoracic Ossification of the Ligamentum Flavum in Chinese Population
Jialiang Lin,Fei Xu,Shuai Jiang,Longjie Wang,Zhuoran Sun,Zhongqiang Chen,Zhaoqing Guo,Qiang Qi,Yan Zeng,Chuiguo Sun,Weishi Li
DOI: https://doi.org/10.1007/s00586-022-07362-0
IF: 2.721
2022-01-01
European Spine Journal
Abstract:To investigate the risk factors for thoracic ossification of the ligamentum flavum (TOLF), especially the relationship between BMI and TOLF. A total of 856 individuals consisting of 326 controls without ossification of spinal ligaments and 530 TOLF inpatients who underwent thoracic spine decompression surgery at our hospital between January 2013 and September 2020 were included. All subjects were classified into 4 grades: Grade 0) control; Grade 1) single-segment TOLF; Grade 2) multi-segment TOLF; and Grade 3) TOLF combined thoracic ossification of the posterior longitudinal ligament (T-OPLL). Logistic regression analysis was performed to identify the risk factors for TOLF. The TOLF index was calculated to assess the severity of TOLF, and its relationship with BMI was investigated by correlation analysis. Overall, TOLF patients are most numerous in the 50–59 age group. Age and gender were considered as independent risk factors for Grades 1 and 2. BMI was identified as an independent risk factor for TOLF. Furthermore, BMI was significantly higher in Grade 1 (26.1 VS 24.5 kg/m2, P = 0.0001), Grade 2 (28.2 VS 24.5 kg/m2, P < 0.0001), and Grade 3 (29.1 VS 24.5 kg/m2, P < 0.0001) than Grade 0. Notably, in TOLF patients without combined T-OPLL, BMI was positively correlated with TOLF index, while BMI was negatively correlated with age in younger individuals. BMI is a crucial risk factor for TOLF. It highlights the necessity of close follow-up of asymptomatic TOLF patients with high BMI to detect and treat their TOLF progression promptly.
-
Tissue Transglutaminase Expression and Activity in Human Ligamentum Flavum Cells Derived from Thoracic Ossification of Ligamentum Flavum
Xiaoxue Yin,Zhongqiang Chen,Zhaoqing Guo,Xiaoguang Liu,Haiyan Yu
DOI: https://doi.org/10.1097/brs.0b013e3181e198ab
IF: 3.241
2010-01-01
Spine
Abstract:STUDY DESIGN:The study was undertaken to compare the expression and activity of tissue transglutaminase (TG2) in human ligamentum flavum cells derived from ossification of the ligamentum flavum (OLF) and non-OLF patients.OBJECTIVE:To determine whether TG2 is involved in the pathologic process of OLF.SUMMARY OF BACKGROUND DATA:OLF is a disease characterized by heterotopic formation of new bone in the flavum ligament. Recently, TG2 is proved to directly promote skeletal matrix mineralization and play an important role in the ossification. TG2 activity is vital to the differentiation of osteoblasts and the formation of mineralization. But whether TG2 is involved in the pathologic process of OLF is unknown. We investigated the relations between TG2 expression and OLF.METHODS:OLF and non-OLF cells were cultured and osteocalcin, bone morphogenetic protein-2(BMP-2) and TG2 mRNA expressions were assayed by reverse transcription polymerase chain reaction. Meanwhile, alkaline phosphatase activity and calcified nodules were compared between OLF and non-OLF cells. To detect TG2 expression, Western blot and immunohistochemical analysis were carried out, and TG2 activity was compared between OLF and non-OLF cells.RESULTS:Our experiments demonstrated that OLF cells showed osteoblast-like activity and increased mRNA expression of BMP-2. More interesting, compared with non-OLF cells, OLF cells showed elevated expression levels of TG2 mRNA and protein, as well as enzyme activity.CONCLUSION:TG2 expression and enzyme activity are upregulated in the OLF cells and TG2 may be involved in the pathologic process of OLF.
-
Genetic Differences in Osteogenic Differentiation Potency in the Thoracic Ossification of the Ligamentum Flavum under Cyclic Mechanical Stress
Shanglong Ning,Zhongqiang Chen,Dongwei Fan,Chuiguo Sun,Chi Zhang,Yan Zeng,Weishi Li,Xiaofei Hou,Xiaochen Qu,Yunlong Ma,Huilei Yu
DOI: https://doi.org/10.3892/ijmm.2016.2803
IF: 5.314
2016-01-01
International Journal of Molecular Medicine
Abstract:Mechanical stress and genetic factors play important roles in the occurrence of thoracic ossification of ligament flavum (TOLF), which can occur at one, two, or multiple levels of the spine. It is unclear whether single- and multiple-level TOLF differ in terms of osteogenic differentiation potency and osteogenesis-related gene expression under cyclic mechanical stress. This was addressed in the present study using patients with non-TOLF and single- and multiple-level TOLF (n=8 per group). Primary ligament cells were cultured and osteogenesis was induced by application of cyclic mechanical stress. Osteogenic differentiation was assessed by evaluating alkaline phosphatase (ALP) activity and the mRNA and protein expression of osteogenesis-related genes, including ALP, bone morphogenetic protein 2 (BMP2), Runt-related transcription factor-2 (Runx-2), osterix, osteopontin (OPN) and osteocalcin. The application of cyclic mechanical stress resulted in higher ALP activity in the multiple-level than in the single-level TOLF group, whereas no changes were observed in the non-TOLF group. The ALP, BMP2, OPN and osterix mRNA levels were higher in the multiple-level as compared to the single-level TOLF group, and the levels of all osteogenesis-related genes, apart from Runx2, were higher in the multiple-level as compared to the non-TOLF group. The osterix and ALP protein levels were higher in the multiple-level TOLF group than in the other 2 groups, and were increased with the longer duration of stress. These results highlight the differences in osteogenic differentiation potency between single- and multiple-level TOLF that may be related to the different pathogenesis and genetic background.
-
The Effect of Mechanical Stress on the Osteogenic Differentiation in Thoracic Ossification of Ligamentum Flavum
Shanglong NING,Xinlong MA,Dongwei FAN,Jun MIAO
DOI: https://doi.org/10.13704/j.cnki.jyyx.2017.09.026
2017-01-01
Abstract:Objective To investigate the effect of mechanical stress on the osteogenic differentiation in thoracic ossification of ligamentum flavum.Methods Clinically,the patients with non-TOLF and TOLF (n=10 in each group) were included in this study.The primary ligament cells that derived from the two groups were separately cultured and induced osteogenesis with 15% strength of cyclic mechanical stressfor 24 h to using a device called Flexcell FX-4000.The ALP activity was determined to evaluate the osteogenesis using quantitative analysis and ALP staining assay.Real-time PCR was used to detect the mRNA expression of osteogenic-related genes including ALP,BMP-2,Osterix,0steopontin and Osteocalcin.Results The morphology of the cells that derived from two groups was basically similar,all presented an elongate spindle-shape.To evaluate the ostogenesis,ALP activity assays including quantitative and staining assays were performed.The results showed that either quantitative analysis or ALP staining in the TOLF group was significantly higher than non-TOLF group.The results of real-time PCR indicated that the expression of ostegenic markers above in the TOLF group was more than non-TOLF group.Therein,the significant difference was found in ALP,BMP-2.However,all indicators in the TOLF group were significantly higher after the osteogenic induction for 24 h.Conclusion The mechanical stress is possible to play an important role in the progression of TOLF,which enhance the osteogenic differentiation by affecting some osteogenic genes.