Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Int Endod J ; 57(6): 727-744, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38436622

ABSTRACT

AIMS: This study aimed to investigate the anti-inflammatory and odontoblastic effects of cerium-containing mesoporous bioactive glass nanoparticles (Ce-MBGNs) on dental pulp cells as novel pulp-capping agents. METHODOLOGY: Ce-MBGNs were synthesized using a post-impregnation strategy based on the antioxidant properties of Ce ions and proposed the first use of Ce-MBGNs for pulp-capping application. The biocompatibility of Ce-MBGNs was analysed using the CCK-8 assay and apoptosis detection. Additionally, the reactive oxygen species (ROS) scavenging ability of Ce-MBGNs was measured using the 2,7-Dichlorofuorescin Diacetate (DCFH-DA) probe. The anti-inflammatory effect of Ce-MBGNs on THP-1 cells was further investigated using flow cytometry and quantitative real-time polymerase chain reaction (RT-qPCR). Moreover, the effect of Ce-MBGNs on the odontoblastic differentiation of the dental pulp cells (DPCs) was assessed by combined scratch assays, RT-qPCR, western blotting, immunocytochemistry, Alizarin Red S staining and tissue-nonspecific alkaline phosphatase staining. Analytically, the secretions of tumour necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) were detected with enzyme-linked immunosorbent assay (ELISA). RESULTS: Ce-MBGNs were confirmed to effectively scavenge ROS in THP-1-derived macrophages and DPCs. Flow cytometry and RT-qPCR assays revealed that Ce-MBGNs significantly inhibited the M1 polarization of macrophages (Mφ). Furthermore, the protein levels of TNF-α and IL-1ß were downregulated in THP-1-derived macrophages after stimulation with Ce-MBGNs. With a step-forward virtue of promoting the odontoblastic differentiation of DPCs, we further confirmed that Ce-MBGNs could regulate the formation of a conductive immune microenvironment with respect to tissue repair in DPCs, which was mediated by macrophages. CONCLUSIONS: Ce-MBGNs protected cells from self-produced oxidative damage and exhibited excellent immunomodulatory and odontoblastic differentiation effects on DPCs. As a pulp-capping agent, this novel biomaterial can exert anti-inflammatory effects and promote restorative dentine regeneration in clinical treatment. We believe that this study will stimulate further correlative research on the development of advanced pulp-capping agents.


Subject(s)
Anti-Inflammatory Agents , Cerium , Dental Pulp , Nanoparticles , Dental Pulp/cytology , Dental Pulp/drug effects , Cerium/pharmacology , Humans , Anti-Inflammatory Agents/pharmacology , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ceramics/pharmacology , Cell Differentiation/drug effects , Glass , Odontoblasts/drug effects , Regeneration/drug effects , THP-1 Cells , Pulp Capping and Pulpectomy Agents/pharmacology , Interleukin-1beta/metabolism , Apoptosis/drug effects , Porosity , Cells, Cultured
2.
Int J Biol Sci ; 19(16): 5187-5203, 2023.
Article in English | MEDLINE | ID: mdl-37928259

ABSTRACT

Periodontitis is a highly prevalent chronic inflammatory disease with an exaggerated host immune response, resulting in periodontal tissue destruction and potential tooth loss. The long non-coding RNA, LncR-ANRIL, located on human chromosome 9p21, is recognized as a genetic risk factor for various conditions, including atherosclerosis, periodontitis, diabetes, and cancer. LncR-APDC is an ortholog of ANRIL located on mouse genome chr4. This study aims to comprehend the regulatory role of lncR-APDC in periodontitis progression. Our experimental findings, obtained from lncR-APDC gene knockout (KO) mice with induced experimental periodontitis (EP), revealed exacerbated bone loss and disrupted pro-inflammatory cytokine regulation. Downregulation of osteogenic differentiation occurred in bone marrow stem cells harvested from lncR-APDC-KO mice. Furthermore, single-cell RNA sequencing of periodontitis gingival tissue revealed alterations in the proportion and function of immune cells, including T and B cells, macrophages, and neutrophils, due to lncR-APDC silencing. Our findings also unveiled a previously unidentified epithelial cell subset that is distinctively presenting in the lncR-APDC-KO group. This epithelial subset, characterized by the positive expression of Krt8 and Krt18, engages in interactions with immune cells through a variety of ligand-receptor pairs. The expression of Tff2, now recognized for its role in chronic inflammatory conditions, exhibited a notable increase across various tissue and cell types in lncR-APDC deficient mice. Additionally, our investigation revealed the potential for a direct binding interaction between lncR-APDC and Tff2. Intra-gingival administration of AAV9-lncR-APDC was shown to have therapeutic effects in the EP model. In conclusion, our results suggest that lncR-APDC plays a critical role in the progression of periodontal disease and holds therapeutic potential for periodontitis. Furthermore, the presence of the distinctive epithelial subpopulation and significantly elevated Tff2 levels in the lncR-APDC-silenced EP model offer new perspectives on the epigenetic regulation of periodontitis pathogenesis.


Subject(s)
Periodontitis , RNA, Long Noncoding , Animals , Humans , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Osteogenesis , Epigenesis, Genetic/genetics , Periodontitis/genetics , Periodontitis/metabolism , Periodontitis/pathology , Cytokines/metabolism , Mice, Knockout
3.
RNA Biol ; 20(1): 836-846, 2023 01.
Article in English | MEDLINE | ID: mdl-37953645

ABSTRACT

The long noncoding RNA (lncR) ANRIL in the human genome is an established genetic risk factor for atherosclerosis, periodontitis, diabetes, and cancer. However, the regulatory role of lncR-ANRIL in bone and adipose tissue metabolism remains unclear. To elucidate the function of lncRNA ANRIL in a mouse model, we investigated its ortholog, AK148321 (referred to as lncR-APDC), located on chr4 of the mouse genome, which is hypothesized to have similar biological functions to ANRIL. We initially revealed that lncR-APDC in mouse bone marrow cells (BMSCs) and lncR-ANRIL in human osteoblasts (hFOBs) are both increased during early osteogenesis. Subsequently, we examined the osteogenesis, adipogenesis, osteoclastogenesis function with lncR-APDC deletion/overexpression cell models. In vivo, we compared the phenotypic differences in bone and adipose tissue between APDC-KO and wild-type mice. Our findings demonstrated that lncR-APDC deficiency impaired osteogenesis while promoting adipogenesis and osteoclastogenesis. Conversely, the overexpression of lncR-APDC stimulated osteogenesis, but impaired adipogenesis and osteoclastogenesis. Furthermore, KDM6B was downregulated with lncR-APDC deficiency and upregulated with overexpression. Through binding-site analysis, we identified miR-99a as a potential target of lncR-APDC. The results suggest that lncR-APDC exerts its osteogenic function via miR-99a/KDM6B/Hox pathways. Additionally, osteoclasto-osteogenic imbalance was mediated by lncR-APDC through MAPK/p38 and TLR4/MyD88 activation. These findings highlight the pivotal role of lncR-APDC as a key regulator in bone and fat tissue metabolism. It shows potential therapeutic for addressing imbalances in osteogenesis, adipogenesis, and osteoclastogenesis.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Humans , Mice , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Bone and Bones/metabolism , Osteogenesis/genetics , Adipose Tissue/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Jumonji Domain-Containing Histone Demethylases
4.
J Bone Miner Metab ; 39(6): 962-973, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34191125

ABSTRACT

INTRODUCTION: Corticotomy is widely used in clinical practice to accelerate tooth movement and shorten the duration of orthodontic treatment. It is effective, but an invasive surgery is needed to induce alveolar bone osteopenia that enable rapid tooth movement. In this study, we discovered the potential of 6-shogaol as a more patient-friendly non-invasive alternative to induce transient osteopenia and accelerate tooth movement. MATERIALS AND METHODS: The effects of 6-shogaol on the bone marrow macrophages (BMM) proliferation and osteoclast differentiation, and bone resorption were determined in vitro. Sprague-Dawley rats were distributed into three groups: CON, IPinj or Localinj and euthanized at day 28. Micro-CT, histology, immunohistological, and TUNEL analysis were performed to evaluate the tooth movement acceleration effect of 6-shogaol. RESULTS: In vitro, 6-shogaol promotes osteoclast differentiation and functional demineralization of alveolar bone. RANKL-induced mRNA expression of osteoclastic-specific genes was significantly higher in the presence of 6-shogaol. A dose-dependent increase in the area of TRAP-positive cells was observed with 6-shogaol treatment. F-actin ring formation and increased bone resorption confirmed that osteoclasts treated with 6-shogaol were mature and functional. 6-shogaol stimulated JNK activation and NFATc1 expression during osteoclast differentiation. In vivo, 6-shogaol promotes alveolar bone transient osteopenia and accelerates orthodontic tooth movement. Alveolar bone mass was reduced, more osteoclasts were observed in bone resorption lacunae on the compression side, and the expression of RANKL and sclerostin were higher than the control group. In conclusion, our results suggest that 6-shogoal accelerates tooth movement by inducing osteopenia by a mechanism similar to surgically induced bone injury.


Subject(s)
Bone Resorption , Tooth Movement Techniques , Animals , Catechols , Humans , NFATC Transcription Factors , Osteoclasts , Rats , Rats, Sprague-Dawley
5.
J Cell Mol Med ; 22(4): 2162-2176, 2018 04.
Article in English | MEDLINE | ID: mdl-29377540

ABSTRACT

Skeletal tissue originates from mesenchymal stem cells (MSCs) with differentiation potential into the osteoblast lineage regulated by essential transcriptional and post-transcriptional mechanisms. Recently, miRNAs and histone modifications have been identified as novel key regulators of osteogenic differentiation of MSCs. Here, we identified miR-99a and its target lysine (K)-specific demethylase 6B (KDM6B) gene as novel modulators of osteogenic differentiation of bone mesenchymal stem cells (BMSCs). Microarray profiling and further validation by quantitative real-time RT-PCR revealed that miR-99a was up-regulated during osteoblastic differentiation of BMSCs, and decreased in differentiated osteoblasts. Transfection of miR-99a mimics inhibited osteoblastic commitment and differentiation of BMSCs, whereas inhibition of miR-99a by inhibitors enhances these processes. KDM6B was determined as one of important targets of miR-99a, which was further confirmed by luciferase assay of 3'-UTR of KDM6B. Moreover, HOX gene level decreased after transfection of miR-99a mimics in BMSCs, which indicated that KDM6B is a bona fide target of miR-99a. Furthermore, in a model of in vivo bone regeneration, osteoblast-specific gain- and loss-of-function experiments performed using cranial bone defects revealed that miR-99a mimics-transfected BMSCs reduced bone formation, and conversely, miR-99a inhibitors-transfected BMSCs increased in vivo bone formation. Tissue-specific inhibition of miR-99a may be a potential novel therapeutic approach for enhancing BMSCs-based bone formation and regeneration.


Subject(s)
Cell Differentiation/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Osteogenesis/genetics , Animals , Base Sequence , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Line , Cells, Cultured , Down-Regulation/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Osteoporosis/genetics , Osteoporosis/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , Wound Healing
6.
Biochem Biophys Res Commun ; 316(2): 461-7, 2004 Apr 02.
Article in English | MEDLINE | ID: mdl-15020240

ABSTRACT

Regulation of the bone sialoprotein (BSP) gene is important in the differentiation of osteoblasts, in bone matrix mineralization, and in tumor metastasis. We investigated BSP gene transcription by performing functional analysis of the 9256bp of the 5' flanking region of the murine BSP gene containing its promoter. We found that the forced expression of BSP stimulated mouse BSP promoter activity in a dose-dependent manner in both MC3T3-E1 preosteoblast and HEK-293 cell lines, which was transcriptional factor Cbfa1 independent. Co-culture of cells separately expressing BSP promoter reporter and BSP failed to mediate the BSP autoregulation, suggesting that the event might happen intracellularly. Deletion analysis of the BSP promoter indicated that the proximal promoter (110bp) was sufficient to confer this autoregulation. We conclude that the BSP gene is autoregulated in part by a positive feedback on its own promoter.


Subject(s)
Osteoblasts/metabolism , Sialoglycoproteins/genetics , Stem Cells/metabolism , Transcriptional Activation , Animals , Cell Line , Coculture Techniques , Cytomegalovirus/genetics , Genetic Vectors , Homeostasis , Humans , Integrin-Binding Sialoprotein , Mice , Promoter Regions, Genetic , RNA, Messenger/metabolism , Regulatory Sequences, Nucleic Acid , Sialoglycoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL