Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters

Publication year range
1.
FASEB J ; 36(4): e22231, 2022 04.
Article in English | MEDLINE | ID: mdl-35230719

ABSTRACT

The dysfunction of osteogenesis is a key character in the pathogenesis of osteoporosis, but the network of signaling mechanisms in controlling the differentiation of osteoblast remain unclear. Thrap3Ā has been proved participating in various biological process, especially in the differentiation of stem cells. Here, we demonstrate that Thrap3 could promote osteogenesis through the inhibition of the degradation of Runx2, which is a key molecular structure in early osteoblast differentiation. Furthermore, we found that the osteogenesis enhancing capacity of Thrap3 was caused by physically binding with Sox9, inhibiting the transcriptional activity of Sox9, and then decreasing the decomposition-promoted effect of Sox9 on Runx2. Our data shows that Thrap3 promotes osteoblast differentiation through the Thrap3-Sox9-Runx2 axis. What we found may help for further clarifying the molecular mechanism of osteogenic differentiation and give a new potential therapeutic target for osteoporosis.


Subject(s)
Core Binding Factor Alpha 1 Subunit/physiology , DNA-Binding Proteins/physiology , Osteogenesis/physiology , Transcription Factors/physiology , Animals , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Osteoblasts/cytology , SOX9 Transcription Factor/physiology
2.
J Cell Physiol ; 235(10): 7261-7272, 2020 10.
Article in English | MEDLINE | ID: mdl-32180230

ABSTRACT

Breast cancer stem cells (BCSCs) are competent to initiate tumor formation and growth and refractory to conventional therapies. Consequently BCSCs are implicated in tumor recurrence. Many signaling cascades associated with BCSCs are critical for epithelial-to-mesenchymal transition (EMT). We developed a model system to mechanistically examine BCSCs in basal-like breast cancer using MCF10AT1 FACS sorted for CD24 (negative/low in BCSCs) and CD44 (positive/high in BCSCs). Ingenuity Pathway Analysis comparing RNA-seq on the CD24-/low versus CD24+/high MCF10AT1 indicates that the top activated upstream regulators include TWIST1, TGFƟ1, OCT4, and other factors known to be increased in BCSCs and during EMT. The top inhibited upstream regulators include ESR1, TP63, and FAS. Consistent with our results, many genes previously demonstrated to be regulated by RUNX factors are altered in BCSCs. The RUNX2 interaction network is the top significant pathway altered between CD24-/low and CD24+/high MCF10AT1. RUNX1 is higher in expression at the RNA level than RUNX2. RUNX3 is not expressed. While, human-specific quantitative polymerase chain reaction primers demonstrate that RUNX1 and CDH1 decrease in human MCF10CA1a cells that have grown tumors within the murine mammary fat pad microenvironment, RUNX2 and VIM increase. Treatment with an inhibitor of RUNX binding to CBFƟ for 5 days followed by a 7-day recovery period results in EMT suggesting that loss of RUNX1, rather than increase in RUNX2, is a driver of EMT in early stage breast cancer. Increased understanding of RUNX regulation on BCSCs and EMT will provide novel insight into therapeutic strategies to prevent recurrence.


Subject(s)
Breast Neoplasms/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Neoplastic Stem Cells/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 2 Subunit/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Mice , Mice, SCID , Neoplastic Stem Cells/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Tumor Microenvironment/genetics
3.
J Gene Med ; 22(11): e3244, 2020 11.
Article in English | MEDLINE | ID: mdl-32559818

ABSTRACT

BACKGROUND: For cartilage regeneration, stem cells are a promising cell source; however, even the advances made in the differentiation of stem cells into precursor-differentiated cartilage cells have not been successful with respect to reprograming these cells to achieve complete differentiation and fully functioning cells until now. Previous findings suggest that Runx2 plays a major role in chondrocyte differentiation and maturation. Although targeting Runx2 has enhanced some chondrocyte properties, the adipogenic lineage shift has eventually occurred in these cells. The present study mainly aimed to reveal the mechanism of this adipogenesis. METHODS: To create inducible artificial shRNA-miR expressing vectors, the designed short hairpin RNAs (shRNAs) were inserted into the pri-mir-30 backbone, cloned into lentiviral pLVET-Tet-on, and transducted into mesenchymal stem cells (MSCs). Runx2 gene was silenced in MSCs either for 1 week or 4 weeks and cultured in the chondrogenic medium. At days 7, 14 and 28, cells were harvested, and chondrogenesis, adipogenesis and hypertrophic states were examined using histochemical staining and a real-time polymerase chain reaction assay. RESULTS: The results showed that the designed shRNA-miR effectively targeted Runx2 in mRNA and protein levels. Chondrogenic markers were up-regulated in constantly silenced Runx2 group; however, adipogenic markers and fat droplets appeared gradually. DLK1 gene was also significantly down-regulated in this group, and overexpression of DLK1 abrogated adipogenesis in the Runx2 targeted group. CONCLUSIONS: Based on these results, it can be concluded that DLK1 is responsible for the lineage shift in Runx2 targeted chondrogenic differentiating MSCs.


Subject(s)
Adipogenesis , Calcium-Binding Proteins/antagonists & inhibitors , Cell Differentiation , Chondrocytes/cytology , Chondrogenesis , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Mesenchymal Stem Cells/cytology , Cell Proliferation , Cells, Cultured , Chondrocytes/metabolism , Humans , Mesenchymal Stem Cells/metabolism
4.
Biochem Biophys Res Commun ; 510(1): 53-58, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30660362

ABSTRACT

PURPOSE: The aim of this study was to investigate the mechanism of let-7a-5p in osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in postmenopausal osteoporosis (PMOP) mice. METHODS: A mouse model of PMOP was established and osteoporosis model was identified by micro-CT scan. BMSCs in the sham group and PMOP group were cultured and osteogenic differentiation was induced. The expression of let-7a-5p in BMSCs was detected by qRT-PCR, and BMSCs was induced by osteogenic differentiation in sham and PMOP group. The BMSCs treated by let-7a-5p mimics, let-7a-5p inhibitor and negative control were named as let-7a-5p mimics group, mimics NC group, let-7a-5p inhibitor group and inhibitor NC group, respectively. ALP staining and alizarin red staining were used to detect osteogenic differentiation ability, qRT-PCR and western blot were used to detect the expression of Runt-related transcription factor 2 (Runx2) and Osterix. The targeting relationship between let-7a-5p and TGFBR1 were verificated by target scan and luciferase reporter gene assay. RESULTS: The PMOP mouse model was successfully established. The expression of let-7a-5p in BMSCs of PMOP group was significantly higher than that in the sham group (PĆ¢Ā€ĀÆ<Ć¢Ā€ĀÆ0.05). Let-7a-5p reduced the expression of ALP and the formation of calcified nodules, while also inhibited the expression of Runx2 and Osterix. TGFBR1 is the target gene of let-7a-5p. CONCLUSION: Let-7a-5p might inhibit the osteogenic differentiation of BMSCs in PMOP mice by regulating TGFBR1.


Subject(s)
Mesenchymal Stem Cells/cytology , MicroRNAs/physiology , Osteogenesis/drug effects , Osteoporosis, Postmenopausal/prevention & control , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Animals , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Disease Models, Animal , Humans , Mesenchymal Stem Cells/drug effects , Mice , MicroRNAs/pharmacology , Sp7 Transcription Factor/antagonists & inhibitors
5.
Cell Physiol Biochem ; 49(1): 217-225, 2018.
Article in English | MEDLINE | ID: mdl-30138923

ABSTRACT

BACKGROUND/AIMS: Esophageal carcinoma is a frequently occurring cancer at upper gastrointestinal tract. We aimed to evaluate the roles and possible mechanism of Runt Related Transcription Factor 2 (RUNX2) in the development of esophageal cancer. METHODS: The expression of RUNX2 in esophageal carcinoma tissues and cells was investigated by qRT-PCR. Effects of RUNX2 on cell viability, apoptosis, migration and invasion were assessed using MTT assay, flow cytometry assay/western blot analysis, and Transwell assays, respectively. Afterwards, effects of RUNX2 on of the activation of the PI3K/AKT and ERK pathways were explored by Western blot analysis. In addition, a PI3K/AKT pathway inhibitor LY294002 and an ERK inhibitor U0126 were applied to further verify the regulatory relationship between RUNX2 and the PI3K/AKT and ERK signaling pathways. Besides, the RUNX2 function on tumor formation in vivo was investigated by tumor xenograft experiment. RESULTS: The result showed that RUNX2 was highly expressed in esophageal carcinoma tissues and cells. Knockdown of RUNX2 significantly inhibited TE-1 and EC-109 cell viability, repressed TE-1 cell migration and invasion, and increased TE-1 cell apoptosis. RUNX2 overexpression showed the opposite effects on HET-1A cells. Moreover, RUNX2-mediated TE-1 cell viability, migration and invasion were associated with the activation of the PI3K/AKT and ERK pathways. Besides, knockdown of RUNX2 markedly suppressed tumor formation in vivo. CONCLUSION: Our results indicate that RUNX2 may play an oncogenic role in esophageal carcinoma by activating the PI3K/ AKT and ERK pathways. RUNX2 may serve as a potent target for the treatment of esophageal carcinoma.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Esophageal Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Butadienes/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chromones/pharmacology , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/genetics , Esophageal Neoplasms/metabolism , Female , Humans , Male , Mice , Mice, Nude , Middle Aged , Morpholines/pharmacology , Nitriles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects
6.
Bioorg Med Chem Lett ; 27(21): 4789-4793, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29017788

ABSTRACT

Zaluzanin C (ZC) is a sesquiterpene lactones used in herbal medicines. This study examined the effects of ZC on osteoblast differentiation. ZC-induced mRNA expressions levels of osteogenic genes in C3H10T1/2 and MC3T3-E1 cells were determined by RT-PCR and qPCR. ZC regulated the expression of key osteogenic genes in the early stage of differentiation, including distal-less homeobox 5 (Dlx5), DNA-binding protein inhibitor (Id1) and Runt-related transcription factor 2 (Runx2). In addition, ZC increased Runx2 promoter activity, as assessed via a luciferase assay, and Runx2 protein level. These results suggest that ZC may enhance osteoblast differentiation by upregulating the expression of osteogenic genes, especially early stage like as Dlx5, Id1 and Runx2.


Subject(s)
Cell Differentiation/drug effects , Gene Expression/drug effects , Osteogenesis/drug effects , Sesquiterpenes, Guaiane/pharmacology , Alkaline Phosphatase/metabolism , Animals , Cell Line , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism
7.
J Immunol ; 195(1): 217-26, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26019273

ABSTRACT

Proper regulation of microbial-induced cytokines is critical to intestinal immune homeostasis. Acute stimulation of nucleotide-binding oligomerization domain 2 (NOD2), the Crohn's disease-associated sensor of bacterial peptidoglycan, induces cytokines. However, chronic NOD2 stimulation in macrophages decreases cytokines upon pattern recognition receptor (PRR) restimulation; cytokine attenuation to PRR stimulation is similarly observed in intestinal macrophages. The role for the transcriptional repressors Twist1 and Twist2 in regulating PRR-induced cytokine outcomes is poorly understood and has not been reported for NOD2. We found that Twist1 and Twist2 were required for optimal cytokine downregulation during acute and, particularly, chronic NOD2 stimulation of human macrophages. Consistently, Twist1 and Twist2 expression was increased after chronic NOD2 stimulation; this increased expression was IL-10 and TGF-Ɵ dependent. Although Twist1 and Twist2 did not coregulate each other's expression, they cooperated to enhance binding to cytokine promoters after chronic NOD2 stimulation. Moreover, Twist1 and Twist2 contributed to enhance expression and promoter binding of the proinflammatory inhibitor c-Maf and the transcriptional repressor Bmi1. Restoring c-Maf and Bmi1 expression in Twist-deficient macrophages restored NOD2-induced cytokine downregulation. Furthermore, with chronic NOD2 stimulation, Twist1 and Twist2 contributed to the decreased expression and cytokine promoter binding of the transcriptional activators activating transcription factor 4, C/EBPα, Runx1, and Runx2. Knockdown of these transcriptional activators in Twist-deficient macrophages restored cytokine downregulation after chronic NOD2 stimulation. Finally, NOD2 synergized with additional PRRs to increase Twist1 and Twist2 expression and Twist-dependent pathways. Therefore, after chronic NOD2 stimulation Twist1 and Twist2 coordinate the regulation of both transcriptional activators and repressors, thereby mediating optimal cytokine downregulation.


Subject(s)
Macrophage Activation/drug effects , Macrophages/immunology , Nod2 Signaling Adaptor Protein/immunology , Nuclear Proteins/immunology , Repressor Proteins/immunology , Twist-Related Protein 1/immunology , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Activating Transcription Factor 4/antagonists & inhibitors , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/immunology , Antibodies, Neutralizing/pharmacology , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/immunology , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/immunology , Gene Expression Regulation , Humans , Interleukin-10/antagonists & inhibitors , Interleukin-10/genetics , Interleukin-10/immunology , Macrophages/cytology , Macrophages/drug effects , Nod2 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/genetics , Nuclear Proteins/genetics , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/immunology , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Repressor Proteins/genetics , Signal Transduction , Transcription, Genetic , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Twist-Related Protein 1/genetics
8.
Lipids Health Dis ; 16(1): 201, 2017 Oct 13.
Article in English | MEDLINE | ID: mdl-29029615

ABSTRACT

BACKGROUND: This study is to investigate the effect of fenofibrate on the bone quality of Type 2 diabetes mellitus (T2DM) mouse model. METHODS: T2DM mouse model was induced by high-fat-diet, and the mice were treated with fenofibrate (100Ā mg/kg) (DIO-FENO) or PBS (DIO-PBS) for 4Ā weeks. The bone microstructure and biomechanical properties of femora were analyzed by micro-CT and 3-Point bending test. The protein expression was detected by immunohistochemical staining and Western blot. The cell apoptosis was evaluated by TUNEL staining. The Bcl2, caspase 3, and osteoblast marker genes were detected by RT-qPCR. RESULTS: The biomechanical properties of bones from DIO-FENO group were significantly lower than those in the control and DIO-PBS groups. Besides, the trabecular number was lower than those of the other groups, though the cortical porosity was decreased compared with that of DIO-PBS group because of the increase of apoptotic cells. The expression of osteocalcin and collagen I were decreased after treatment with fenofibrate in T2DM mice. Moreover, the cell viability was decreased after treated with different concentrations of fenofibrate, and the expression of Runx2 decreased after treated with high dose of fenofibrate. CONCLUSION: Fenofibrate decreases the bone quality of T2DM mice through decreasing the expression of collagen I and osteocalcin, which may be resulted from the down regulation of Runx2 expression.


Subject(s)
Bone Density/drug effects , Core Binding Factor Alpha 1 Subunit/genetics , Diabetes Mellitus, Type 2/genetics , Dyslipidemias/genetics , Fenofibrate/adverse effects , Hypolipidemic Agents/adverse effects , Animals , Caspase 3/genetics , Caspase 3/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/metabolism , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat/adverse effects , Disease Models, Animal , Dyslipidemias/etiology , Dyslipidemias/metabolism , Dyslipidemias/pathology , Gene Expression Regulation , Humans , Mice , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Osteocalcin/genetics , Osteocalcin/metabolism , Porosity/drug effects , Primary Cell Culture , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , X-Ray Microtomography
9.
J Cell Mol Med ; 20(4): 678-87, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26818776

ABSTRACT

ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family is known to play an important role in the pathogenesis of osteoarthritis (OA), working on aggrecan degradation or altering the integrity of extracellular matrix (ECM). Thus, the main purpose of our study was to define the role of vasoactive intestinal peptide (VIP) and corticotrophin-releasing factor (CRF), as immunoregulatory neuropeptides, on ADAMTS production in synovial fibroblasts (SF) from OA patients and healthy donors (HD). OA- and HD-SF were stimulated with pro-inflammatory mediators and treated with VIP or CRF. Both neuropeptides decreased ADAMTS-4, -5, -7 and -12 expressions, aggrecanase activity, glycosaminoglycans (GAG), and cartilage oligomeric matrix protein (COMP) degradation after stimulation with fibronectin fragments (Fn-fs) in OA-SF. After stimulation with interleukin-1Ɵ, VIP reduced ADAMTS-4 and -5, and both neuropeptides decreased ADAMTS-7 production and COMP degradation. Moreover, VIP and CRF reduced Runx2 and Ɵ-catenin activation in OA-SF. Our data suggest that the role of VIP and CRF on ADAMTS expression and cartilage degradation could be related to the OA pathology since scarce effects were produced in HD-SF. In addition, their effects might be greater when a degradation loop has been established, given that they were higher after stimulation with Fn-fs. Our results point to novel OA therapies based on the use of neuropeptides, since VIP and CRF are able to stop the first critical step, the loss of cartilage aggrecan and the ECM destabilization during joint degradation.


Subject(s)
ADAMTS Proteins/genetics , Cartilage, Articular/metabolism , Corticotropin-Releasing Hormone/metabolism , Fibroblasts/metabolism , Osteoarthritis/genetics , Vasoactive Intestinal Peptide/metabolism , ADAMTS Proteins/antagonists & inhibitors , ADAMTS Proteins/metabolism , Aged , Aged, 80 and over , Cartilage Oligomeric Matrix Protein/genetics , Cartilage Oligomeric Matrix Protein/metabolism , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Case-Control Studies , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Corticotropin-Releasing Hormone/pharmacology , Endopeptidases/genetics , Endopeptidases/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Fibronectins/pharmacology , Gene Expression Regulation , Glycosaminoglycans/metabolism , Humans , Interleukin-1beta/pharmacology , Joint Capsule/metabolism , Joint Capsule/pathology , Male , Middle Aged , Osteoarthritis/metabolism , Osteoarthritis/pathology , Signal Transduction , Vasoactive Intestinal Peptide/pharmacology , beta Catenin/antagonists & inhibitors , beta Catenin/genetics , beta Catenin/metabolism
10.
J Cell Physiol ; 231(2): 473-82, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26189652

ABSTRACT

Aberrant expression of the transcription factor RUNX2 in prostate cancer has a number of important consequences including increased resistance to apoptosis, invasion and metastasis to bone. We previously demonstrated that hypoxia up-regulated RUNX2 in tumour cells, which in turn up-regulated the anti-apoptotic factor Bcl-2. Here, we investigate the impact of nitric oxide (NO) on RUNX2 and Bcl-2 expression in prostate cancer and further, how RUNX2 over-expression can impact tumour growth, angiogenesis and oxygenation in vivo. The effect of NO levels on RUNX2 and thus Bcl-2 expression was examined in prostate cancer cells in vitro using methods including gene and protein expression analyses, nitrite quantitation, protein-DNA interaction assays (ChIP) and viability assays (XTT). The effect of RUNX2 over-expression on tumour physiology (growth, oxygenation and angiogenesis) was also assessed in vivo using LNCaP xenografts. A low (but not high) concentration of NO (10 ĀµM) induced expression of RUNX2 and Bcl-2, conferring resistance to docetaxel. These effects were induced via the ERK and PI3K pathways and were dependent on intact AP-1 binding sites in the RUNX2 promoter. RUNX2 over-expression in LNCaP tumours in vivo decreased the time to tumour presentation and increased tumour growth. Moreover, these tumours exhibited improved tumour angiogenesis and oxygenation. Low levels of NO increase expression of RUNX2 and Bcl-2 in LNCaP prostate tumour cells, and in vivo up-regulation of RUNX2 created tumours with a more malignant phenotype. Collectively, our data reveals the importance of NO-regulation of key factors in prostate cancer disease progression.


Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Nitric Oxide/metabolism , Prostatic Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Female , Gene Knockdown Techniques , Heterografts , Humans , MCF-7 Cells , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics , Up-Regulation
11.
Mol Hum Reprod ; 22(2): 119-29, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26660506

ABSTRACT

STUDY HYPOTHESIS: We hypothesized that Runt-related transcription factor 2 (RUNX2), matrix metalloproteinase (MMP)2 and MMP9 are involved in basal and gonadotrophin-releasing hormone (GnRH)-induced human extravillous trophoblast (EVT) cell invasion. STUDY FINDING: Our finding indicates that GnRH-induced RUNX2 expression enhances the invasive capacity of EVT cells by modulating the expression of MMP2 and MMP9. WHAT IS KNOWN ALREADY: GnRH is expressed in first-trimester placenta and exerts pro-invasive effects on EVT cells in vitro. RUNX2 regulates MMP2 and MMP9 expression and is often associated with invasive phenotypes. STUDY DESIGN, SAMPLES/MATERIALS, METHODS: First-trimester human placenta (n = 9) was obtained from women undergoing elective termination of pregnancy. The localization of RUNX2, MMP2 and MMP9 in first-trimester human placenta was examined by immunohistochemistry. Primary or immortalized (HTR-8/SVneo) EVT cells were treated alone or in combination with GnRH, GnRH antagonist Antide, MAPK kinase inhibitor PD98095, phosphatidylinositol 3-kinase inhibitor LY294002, MMP2/9 inhibitor or small interfering RNAs (siRNAs) targeting RUNX2, MMP2 and/or MMP9. Protein and mRNA levels were measured by western blot and RT-PCR, respectively. Cell invasiveness was evaluated by transwell Matrigel or collagen I invasion assays. MAIN RESULTS AND THE ROLE OF CHANCE: RUNX2, MMP2 and MMP9 were detected in the cell column regions of human first-trimester placental villi. GnRH treatment increased RUNX2 mRNA and protein levels in HTR-8/SVneo cells and primary EVTs, and these effects were attenuated by co-treatment with Antide, PD98095 or LY294002. Down-regulation of RUNX2 by siRNA reduced basal and GnRH-induced MMP2/9 expression and cell invasion. Moreover, pharmacological inhibition or siRNA-mediated knockdown of MMP2/9 reduced basal and GnRH-induced cell invasion. LIMITATIONS, REASONS FOR CAUTION: The lack of an in vivo model is the major limitation of our in vitro study. WIDER IMPLICATIONS OF THE FINDINGS: Our findings provide important insight into the functions of the GnRH - GnRH receptor system in early implantation and placentation. LARGE SCALE DATA: Not applicable. STUDY FUNDING AND COMPETING INTERESTS: This research was supported by Canadian Institutes of Health Research (Grant #143317) to P.C.K.L. The authors have nothing to disclose.


Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , Gonadotropin-Releasing Hormone/pharmacology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , RNA, Messenger/genetics , Trophoblasts/drug effects , Abortion, Legal , Adult , Cell Line, Transformed , Cell Movement/drug effects , Chorionic Villi/metabolism , Chromones/pharmacology , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/metabolism , Decidua/cytology , Decidua/metabolism , Female , Flavonoids/pharmacology , Gene Expression Regulation , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Morpholines/pharmacology , Oligopeptides/pharmacology , Pregnancy , Pregnancy Trimester, First , Primary Cell Culture , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Trophoblasts/cytology , Trophoblasts/metabolism
12.
J Cardiovasc Pharmacol ; 66(1): 63-71, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25806689

ABSTRACT

Osteoblast differentiation of valve interstitial cells (VICs) is a key step in valve calcification, but the molecular mechanisms involved are not fully understood. In this study, we aimed to investigate whether microRNA (miR)-204-regulated VICs differentiation through modulation of runt-related transcription factor 2 (Runx2), a key transcription factor for osteogenesis. Our data demonstrated that miR-204 was markedly downregulated in both human calcified aortic valves and bone morphogenetic protein (BMP)-2-stimulated aortic VICs. In vitro experiments showed that miR-204 acted as a negative regulator of osteogenic differentiation by repressing Runx2 and thereby inhibiting expression of osteoblast-related genes, including alkaline phosphatase and osteocalcin, which were all induced by BMP-2. Luciferase reporter assays validated Runx2 as the direct target of miR-204. Furthermore, increased alkaline phosphatase activity and osteocalcin expression after miR-204 inhibition were abolished by small interfering RNA-mediated silencing of Runx2. Overall, these data suggested miR-204 as a possible molecular switch inhibiting osteoblastic transdifferentiation of human aortic VICs and targeting miR-204 may have therapeutic potential for human aortic valve calcification.


Subject(s)
Aortic Valve/drug effects , Bone Morphogenetic Protein 2/pharmacology , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , MicroRNAs/administration & dosage , Osteoblasts/drug effects , Aged , Aortic Valve/cytology , Aortic Valve/metabolism , Bone Morphogenetic Protein 2/metabolism , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Humans , Male , MicroRNAs/metabolism , Osteoblasts/metabolism , Osteogenesis/drug effects , Osteogenesis/physiology
13.
Genet Mol Res ; 13(3): 6398-410, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-25158258

ABSTRACT

This study was designed to investigate the effect of different concentrations of rifampicin on osteogenic differentiation and proliferation of mesenchymal stem cells (MSCs) in human bone marrow. Rifampicin treatment at 0, 4, 8, 16, 32, 64, and 128 mg/mL was applied throughout the whole process, from stromal cells purified from human bone marrow to differentiated bone cells. The effect of rifampicin on MSC proliferation was determined using the MTT assay. The effect of rifampicin on the expressions of type I collagen (COL1A1), osteopontin/bone Gla protein (OPN/BGP), and alkaline phosphatase (ALP) in human osteoblast cells were determined by real-time polymerase chain reaction, and the expressions of COL1A1, OPN/BGP, and the runt-related transcription factor (RUNX2) were determined by Western blot. Results showed that the proliferation of MSCs was significantly inhibited when the rifampicin concentration exceeded 32 mg/mL. In addition, increased rifampicin concentrations inhibited the formation of calcium nodules, OPN/BGP, and COL1A1 in osteoblasts after 28 days of induction. The RNA expressions of OPN/BGP, COL1A1, and ALP were significantly downregulated compared to those of the control group in osteoblasts after induction. The protein expressions of RUNX2, COL1A1, and OPN/BGP were also significantly downregulated compared to those of the control group after induction. In conclusion, rifampicin at exorbitant concentration exerts adverse effects on the proliferation of MSCs in human bone marrow and the differentiation of osteoblasts.


Subject(s)
Antitubercular Agents/pharmacology , Bone Marrow Cells/drug effects , Gene Expression/drug effects , Mesenchymal Stem Cells/drug effects , Osteoblasts/drug effects , Rifampin/pharmacology , Alkaline Phosphatase/antagonists & inhibitors , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Collagen Type I/antagonists & inhibitors , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Dose-Response Relationship, Drug , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteopontin/antagonists & inhibitors , Osteopontin/genetics , Osteopontin/metabolism , Primary Cell Culture
14.
Biochem Biophys Res Commun ; 432(4): 612-7, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23438432

ABSTRACT

Long noncoding RNAs (lncRNAs) are key regulators of diverse biological processes such as transcriptional regulation, cell growth and differentiation. Previous studies have demonstrated that the lncRNA-ANCR (anti-differentiation ncRNA) is required to maintain the undifferentiated cell state within the epidermis. However, little is known about whether ANCR regulates osteoblast differentiation. In this study, we found that the ANCR expression level is significantly decreased during hFOB1.19 cell differentiation. ANCR-siRNA blocks the expression of endogenous ANCR, resulting in osteoblast differentiation, whereas ANCR overexpression is sufficient to inhibit osteoblast differentiation. We further demonstrated that ANCR is associated with enhancer of zeste homolog 2 (EZH2) and that this association results in the inhibition of both Runx2 expression and subsequent osteoblast differentiation. These data suggest that ANCR is an essential mediator of osteoblast differentiation, thus offering a new target for the development of therapeutic agents to treat bone diseases.


Subject(s)
Cell Differentiation/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Gene Expression Regulation , Osteoblasts/cytology , Polycomb Repressive Complex 2/genetics , RNA, Long Noncoding/metabolism , Cell Line , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Down-Regulation , Enhancer Elements, Genetic , Enhancer of Zeste Homolog 2 Protein , Humans , Promoter Regions, Genetic , RNA, Long Noncoding/genetics
15.
J Biol Chem ; 285(47): 36369-76, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20843790

ABSTRACT

Dentin sialophosphoprotein (DSPP), a typical dentin-specific protein, is mainly expressed in the dentin extracellular matrix and plays a role in dentin mineralization. BMP-2 provides a strong signal for differentiation and mineralization of odontoblasts and osteoblasts. Previously, BMP-2 treatment is reported to stimulate Dspp expression in the MD10-F2 pre-odontoblast cells through activation of the heterotrimeric transcription factor Y (NF-Y). The canonical BMP signaling pathway is known to contribute greatly to biomineralization, however, it is not known whether it is involved in Dspp expression. Here, we investigated this question. Activation of the canonical BMP-2 signaling pathway in MDPC-23, preodontoblast cell, by overexpression of constitutively active Smad1/5 or downstream transcription factors Dlx5 and Runx2 stimulated Dspp expression. Conversely, knockdown of each element with siRNA significantly blocked the BMP-2-induced Dspp expression. To test whether these transcription factors downstream of BMP-2 are directly involved in regulating Dspp, we analyzed the mouse Dspp promoter. There are 5 well conserved homeodomain binding elements, H1 to H5, in Dspp proximal promoter regions (-791 to +54). A serial deletion of H1 and H2 greatly changed basal promoter activity and responsiveness to Dlx5 or Msx2. However, further deletions did not change the responsiveness to Dlx5 or Msx2. H1 and H2 sites can be suggested as specific response elements of Dlx5 and Msx2, respectively, based on their promoter activity modulation. Thus, the canonical BMP-2 signaling pathway plays a crucial part in the regulation of Dspp expression through the action of Smads, Dlx5, Runx2, and Msx2.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Extracellular Matrix Proteins/genetics , Gene Expression Regulation , Odontoblasts/metabolism , Phosphoproteins/genetics , Response Elements/genetics , Sialoglycoproteins/genetics , Signal Transduction , Animals , Blotting, Western , Bone Morphogenetic Protein 2/genetics , Cell Differentiation , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Electrophoretic Mobility Shift Assay , Extracellular Matrix Proteins/metabolism , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Luciferases/metabolism , Mice , Odontoblasts/cytology , Phosphoproteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Sialoglycoproteins/metabolism , Smad2 Protein/antagonists & inhibitors , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/antagonists & inhibitors , Smad3 Protein/genetics , Smad3 Protein/metabolism
16.
Biochem Biophys Res Commun ; 410(3): 698-704, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21703228

ABSTRACT

Runx2, of the Runx family, is an essential transcription factor that controls bone and tooth development by regulating osteoblast and odontoblast differentiation. However, the function of Runx2 in late stage odontoblast differentiation is not clear. We studied the function of Runx2 in dentinogenesis by generating transgenic mice expressing Runx2 specifically in odontoblasts. We observed dentin formation in postnatal day 3 (P3), P7 and P28 mice and measured the expression levels of Runx2 and matrix proteins in dentin. The odontoblasts in transgenic mice (Tg) lost their tall columnar shape and polarization and dentinal tubules were absent. The dental pulp chamber was dramatically enlarged and the dentin in Tg mice was thinner. Osteoblast-like cells were seen instead of normal odontoblasts and were embedded in a bone-like matrix, indicating that dentin formation was replaced with bone. Predentin was disorganized possessing lacunae that contained odontoblasts. The mandibular molars of Tg mice showed noticeable defects by Micro-CT. Using quantitative real-time PCR, the expression of dentin matrix proteins, particularly dentin sialophosphoprotein (DSPP), was found to be upregulated in 3-day-old Tg mice and downregulated at 1 month of age. These findings indicate that Runx2 inhibited odontoblast terminal differentiation and induced transdifferentiation of odontoblasts to osteoblasts at the late cell differentiation stage. Therefore, Runx2 should be inhibited in odontoblasts to encourage normal cell maturation, differentiation and dentinogenesis.


Subject(s)
Cell Differentiation/genetics , Core Binding Factor Alpha 1 Subunit/physiology , Dentin/growth & development , Dentinogenesis/genetics , Odontoblasts/cytology , Animals , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/genetics , Mice , Mice, Transgenic , Molar/growth & development , Molar/metabolism , Molar/ultrastructure
17.
Adv Dent Res ; 23(3): 280-4, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21677079

ABSTRACT

Stem cells derived from the dental pulp of extracted human third molars (DPSCs) have the potential to differentiate into odontoblasts, osteoblasts, adipocytes, and neural cells when provided with the appropriate conditions. To advance the use of DPSCs for dentin regeneration, it is important to replicate the permissive signals that drive terminal events in odontoblast differentiation during tooth development. Such a strategy is likely to restore a dentin matrix that more resembles the tubular nature of primary dentin. Due to the limitations of culture conditions, the use of ex vivo gene therapy to drive the terminal differentiation of mineralizing cells holds considerable promise. In these studies, we asked whether the forced expression of TWIST1 in DPSCs could alter the potential of these cells to differentiate into odontoblast-like cells. Since the partnership between Runx2 and Twist1 proteins is known to control the onset of osteoblast terminal differentiation, we hypothesized that these genes act to control lineage determination of DPSCs. For the first time, our results showed that Twist1 overexpression in DPSCs enhanced the expression of DSPP, a gene that marks odontoblast terminal differentiation. Furthermore, co-transfection assays showed that Twist1 stimulates Dspp promoter activity by antagonizing Runx2 function in 293FT cells. Analysis of our in vitro data, taken together, suggests that lineage specification of DPSCs can be modulated through ex vivo gene modifications.


Subject(s)
Dental Pulp/cytology , Nuclear Proteins/genetics , Odontoblasts/physiology , Stem Cells/physiology , Twist-Related Protein 1/genetics , Alkaline Phosphatase/analysis , Calcification, Physiologic/genetics , Cell Differentiation/genetics , Cell Line , Cell Lineage/genetics , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/genetics , Exons/genetics , Extracellular Matrix Proteins/analysis , Extracellular Matrix Proteins/genetics , Gene Expression Regulation/genetics , Gene Silencing , Genetic Vectors/genetics , Humans , Introns/genetics , Lentivirus/genetics , Osteoblasts/physiology , Osteocalcin/analysis , Osteopontin/analysis , Phosphoproteins/analysis , Phosphoproteins/genetics , Promoter Regions, Genetic/genetics , RNA, Small Interfering , Sialoglycoproteins/analysis , Sialoglycoproteins/genetics , Transfection/methods
18.
J Healthc Eng ; 2021: 6974225, 2021.
Article in English | MEDLINE | ID: mdl-34257854

ABSTRACT

Prostatic cancer (PCa) is a prevalent form of malignancy based on its high associated levels of mortality and morbidity across the world. MicroRNAs (miRNAs) are significant in the advancement of prostatic cancer. The current study is aimed at exploring the potential roles of miR-373 in PCa. In turn, the study conducted a qRT-PCR test to determine the levels of mRNA. A western blot test was also executed in determining the protein level. The processes of transwell assay and wound healing were integrated in the detection of the potential for PCa cells to invade and migrate. The integration of dual luciferase reporter assay is critical in determining the levels of luciferase activity among prostatic cancer cells. Then, the results showed a net decrease of miR-373 within prostatic cancer cells and tissues. Upregulated miR-373 reduced the invasion and migration potential of PCa cells. Moreover, overexpressed miR-373 increased the levels of E-cadherin and FSP1 as epithelial cell markers. Similarly, the overregulation of miR-373 brought about the upregulation of mesenchymal markers (N-cadherin, Snail, and vimentin). The study predicted runt-related transcription factor 2 (RUNX2) to be a target of miR-373. The luciferase activity of PCa cells was decreased after the cotransfection with miR-373 mimics and RUNX2 3' untranslated region (3'UTR) wild type (WT). Moreover, RUNX2 became upregulated in PCa cells and tissues. The upregulation of miR-373 decreased the mRNA and protein level of RUNX2. However, overexpressed RUNX2 abated the roles of miR-373 in the intrusion and migration of PCa cells and in regulating the expression of epithelial cell markers and mesenchymal markers. In short, miR-373 may regulate the EMT of PCa cells via targeting RUNX2. The miR-373/RUNX2 axis provides a therapeutic target for PCa.


Subject(s)
Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , MicroRNAs , Prostatic Neoplasms , Cell Line, Tumor , Cell Movement/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
19.
Gene Ther ; 17(3): 370-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19940863

ABSTRACT

Heterotopic ossification of muscles, tendons and ligaments are a common problem affecting patient with trauma or received elective surgery. But the existing preventive or therapeutic methods all have disadvantages. Runt-related protein 2 (Runx2) and Smad4 are two regulators that have important roles in the differentiation of osteoblast. In this study, we attempted to examine the effect of Runx2 and Smad4 on the development of heterotopic ossification in vitro. We constructed non-virus-containing small interference RNAs (siRNAs) against Runx2 and Smad4 and tested it with reverse transcriptase-PCR and western blot. We then analyzed the independent effect of Runx2- and Smad4-specific siRNAs and their cooperative effect on the formation of heterotopic ossification induced by trauma in rats. The effects were measured with computed tomography scanning, hematoxylin and eosin staining and immunohistochemistry. We found that the Runx2- and Smad4-specific siRNAs inhibited the expression of Runx2 and Smad4 at the level of messenger RNA and protein. Runx2 and Smad4 independently inhibited the formation of heterotopic ossification. Moreover, their co-expression significantly enhanced the inhibition of heterotopic ossification compared with the independent effect. We suggest that gene therapy to inhibit Runx2 and Smad4 by RNAi could be a powerful approach to prevent or treat heterotopic ossification.


Subject(s)
Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Genetic Therapy/methods , Ossification, Heterotopic/therapy , RNA, Small Interfering/genetics , Smad4 Protein/antagonists & inhibitors , Achilles Tendon , Animals , Core Binding Factor Alpha 1 Subunit/genetics , Gene Transfer Techniques , Male , Ossification, Heterotopic/prevention & control , Rats , Smad4 Protein/genetics
20.
Biochem Biophys Res Commun ; 402(4): 755-61, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-21036155

ABSTRACT

Osteoblasts express Toll like receptor (TLR) 4 and produce osteoclast-activating cytokines in response to the stimulation by lipopolysaccharide (LPS). It has recently been reported that LPS exerts an inhibitory effect on osteoblast differentiation into osteocytes. However, the molecular mechanisms of this inhibitory effect remain ambiguous. The downstream signals of TLR4 are mediated by adaptor molecules including myeloid differentiation factor 88 (MyD88), leading to the activation of mitogen-activated protein kinases (MAPKs), such as extracellular signal-regulated kinases (ERKs), whose activation by LPS requires the upstream serine/threonine kinase, Cot/Tpl2. To determine the signal molecules responsible for the inhibitory effects of LPS on osteoblast differentiation, we examined the in vitro differentiation of the primary osteoblasts from myd88(-/-) and cot/tpl2(-/-) mice. The matrix mineralization by the wild-type and cot/tpl2(-/-) osteoblasts was significantly inhibited by LPS, whereas that of myd88(-/-) was not affected. During differentiation, LPS suppressed the mRNA expression of runt related transcription factor 2 (Runx2), osterix (Sp7), and activating transcription factor 4 (ATF4) in the wild-type, but not in the myd88(-/-) osteoblasts. The inhibitory effect of LPS on the mRNA expression of these transcription factors was absent in the early phase but partially impaired in the late phase of differentiation in the cot/tpl2(-/-) osteoblasts. Thus, the inhibitory effect of LPS on osteoblast differentiation is Myd88-dependent, whereas the degree of its requirement for Cot/Tpl2 varies depending on the differentiation phase.


Subject(s)
Cell Differentiation , Lipopolysaccharides/metabolism , MAP Kinase Kinase Kinases/metabolism , Myeloid Differentiation Factor 88/metabolism , Osteoblasts/physiology , Osteocytes/cytology , Osteogenesis , Proto-Oncogene Proteins/metabolism , Activating Transcription Factor 4/antagonists & inhibitors , Activating Transcription Factor 4/genetics , Animals , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/genetics , Lipopolysaccharides/pharmacology , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Osteoblasts/drug effects , Proto-Oncogene Proteins/genetics , Sp7 Transcription Factor , Toll-Like Receptor 4/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL