Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
J Biomater Appl ; 38(2): 280-291, 2023 08.
Article in English | MEDLINE | ID: mdl-37485690

ABSTRACT

Pulp-Dentin regeneration is a key aspect of maintain tooth vitality and enabling good oral-systemic health. This study aimed to investigate a nanofibrous scaffold loaded with a small molecule i.e. Tideglusib to promote odontogenic differentiation. Tideglusib (GSK-3ß inhibitor) interaction with GSK-3ß was determined using molecular docking and stabilization of ß-catenin was examined by confocal microscopy. 3D nanofibrous scaffolds were fabricated through electrospinning and their physicochemical characterizations were performed. Scaffolds were seeded with mesenchymal stem cells or pre-odontoblast cells to determine the cells proliferation and odontogenic differentiation. Our results showed that Tideglusib (TG) binds with GSK-3ß at Cys199 residue. Stabilization and nuclear translocation of ß-catenin was increased in the odontoblast cells treated with TG. SEM analysis revealed that nanofibers exhibited controlled architectural features that effectively mimicked the natural ECM. UV-Vis spectroscopy demonstrated that TG was incorporated successfully and released in a controlled manner. Both kinds of biomimetic nanofibrous matrices (PCLF-TG100, PCLF-TG1000) significantly stimulated cells proliferation. Furthermore, these scaffolds significantly induced dentinogenic markers (ALP, and DSPP) expression and biomineralization. In contrast to current pulp capping material driving dentin repair, the sophisticated, polymeric scaffold systems with soluble and insoluble spatiotemporal cues described here can direct stem cell differentiation and dentin regeneration. Hence, bioactive small molecule-incorporated nanofibrous scaffold suggests an innovative clinical tool for dentin tissue engineering.


Subject(s)
Nanofibers , Tissue Scaffolds , Tissue Scaffolds/chemistry , Nanofibers/chemistry , beta Catenin , Glycogen Synthase Kinase 3 beta/pharmacology , Molecular Docking Simulation , Cells, Cultured , Cell Differentiation , Tissue Engineering , Dental Pulp
2.
Mater Today Bio ; 17: 100479, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36388451

ABSTRACT

Cell differentiation with the proper three-dimensional (3-D) structure is critical for cells to carry out their cellular functions in tissues. Odontoblasts derived from neural crest cells are elongated and polarized with the cell process, which is decisive for one directional tubular dentin formation. Here, we report that the fibrous topography of scaffolds directs odontoblast-lineage cells to differentiate to have the 3-D structure of odontoblasts through an altered responsiveness to Wnt family member 5A (Wnt5a). In a pulp-exposure animal model, the scaffolds with the nanofibrous topography supported the regeneration of tubular dentin with odontoblast processes. In cultures of pre-odontoblast cells, the nanofibrous topography heightened the cells on the z-axis. The cells on nanofibrous substrate (FIBER) formed stress fiber cytoskeletons on a conventional tissue culture plate (TCP). Differential activation of Cell division control protein 42 (Cdc42) on FIBER and Ras homolog family member A (RhoA) on TCP led to these differences. The signal from Wnt5a-Cdc42 in the cells on FIBER mediated the phosphorylation of JNK and the polarity growth signaling. Taken together, the nanofibrous topography of the scaffolds led to the 3-D structural differentiation of odontoblasts in vitro and in vivo, implying its application for dentin regeneration. Furthermore, the results on the altered activation of Cdc42 by Wnt5a on FIBER provide evidence that the topography of the scaffolds can cause a distinctive cell responsiveness to their micro-environments.

3.
Clin Oral Investig ; 26(3): 2607-2618, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34677694

ABSTRACT

OBJECTIVE: This study aimed to investigate the effect of small molecules incorporated into the engineered nanofibrous scaffold to enhance the osteoblast differentiation MATERIALS AND METHODS: Poly-ε-caprolactone (PCL) nanofiber matrices with lithium chloride (LiCl) were fabricated using the electrospinning technique. Scaffolds were characterized using scanning electron microscopy (SEM) and energy-dispersive X-ray (EDX). Scaffolds were seeded with MC3T3-E1 cells and assessed using Western blots (ß-catenin), alamarBlue assay (proliferation), qPCR (osteoblast differentiation), and mineralization (Alizarin Red staining). RESULTS: We observed LiCl nanofiber scaffolds induced concentration-dependent cell proliferation that correlated with an increased ß-catenin expression indicating sustained Wnt signaling. Next, we examined osteoblast differentiation markers such as osteocalcin (OCN) and Runt-related transcription factor 2 (Runx2) and noted increased expression in LiCl nanofiber scaffolds. We also noted increased bone morphogenetic protein (BMP-2, 4, and 7) expressions suggesting activated Wnt can promote cures to further osteogenic differentiation. Finally, Alizarin Red staining demonstrated increased mineral deposition in LiCl-incorporated nanofiber scaffolds. CONCLUSIONS: Together, these results indicated that LiCl-incorporated nanofiber scaffolds enhance osteoblast differentiation. CLINICAL RELEVANCE: Small molecule-incorporated nanofibrous scaffolds are an innovative clinical tool for bone tissue engineering.


Subject(s)
Nanofibers , Osteogenesis , Cell Differentiation , Cell Proliferation , Osteoblasts , Polyesters/pharmacology , Tissue Engineering/methods , Tissue Scaffolds
4.
ACS Nano ; 14(10): 13217-13231, 2020 10 27.
Article in English | MEDLINE | ID: mdl-32969647

ABSTRACT

Male infertility is a multifactorial condition. Unexplained male infertility is often caused by spermatogenesis dysfunction. Knockout of Pin1, an important regulator of cell proliferation and differentiation, produces male infertility phenotypes such as testicular immaturity and azoospermia with spermatogonia depletion and blood-testis barrier (BTB) dysfunction. Gene therapy has been clinically considered for the treatment of male infertility, but it is not preferred because of the risks of adverse effects in germ cells. Direct intracellular protein delivery using nanoparticles is considered an effective alternative to gene therapy; however, in vivo testicular protein delivery remains a pressing challenge. Here, we investigated the direct intracellular protein delivery strategy using a fibroin nanoparticle-encapsulated cationic lipid complex (Fibroplex) to restore intratesticular PIN1. Local intratesticular delivery of PIN1 via Fibroplex in Pin1 knockout testes produced fertile mice, achieving recovery from the infertile phenotypes. Mechanistically, PIN1-loaded Fibroplex was successfully delivered into testicular cells, including spermatogonial cells and Sertoli cells, and the sustained release of PIN1 restored the gene expression required for the proliferation of spermatogonial cells and BTB integrity in Pin1 knockout testes. Collectively, testicular PIN1 protein delivery using Fibroplex might be an effective strategy for treating male infertility.


Subject(s)
Fibroins , Infertility, Male , Nanoparticles , Animals , Humans , Infertility, Male/drug therapy , Lipids , Male , Mice , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase
5.
J Korean Assoc Oral Maxillofac Surg ; 46(4): 240-249, 2020 Aug 31.
Article in English | MEDLINE | ID: mdl-32855371

ABSTRACT

OBJECTIVES: Although the side effects of radiation therapy vary from mucositis to osteomyelitis depending on the dose of radiation therapy, to date, an experimental animal model has not yet been proposed. The aim of this study was to develop an animal model for assessing complications of irradiated bone, especially to quantify the dose of radiation needed to develop a rat model. MATERIALS AND METHODS: Sixteen Sprague-Dawley rats aged seven weeks with a mean weight of 267.59 g were used. Atraumatic extraction of a right mandibular first molar was performed. At one week after the extraction, the rats were randomized into four groups and received a single dose of external radiation administered to the right lower jaw at a level of 14, 16, 18, or 20 Gy, respectively. Clinical alopecia with body weight changes were compared and bony volumetric analysis with micro-computed tomography (CT), histologic analysis with H&E were performed. RESULTS: The progression of the skin alopecia was different depending on the irradiation dose. Micro-CT parameters including bone volume, bone volume/tissue volume, bone mineral density, and trabecular spaces, showed no significant differences. The progression of osteoradionecrosis (ORN) along with that of inflammation, fibrosis, and bone resorption, was found with increased osteoclast or fibrosis in the radiated group. As the radiation dose increases, osteoclast numbers begin to decrease and osteoclast tends to increase. Osteoclasts respond more sensitively to the radiation dose, and osteoblasts are degraded at doses above 18 Gy. CONCLUSION: A standardized animal model clinically comparable to ORN of the jaw is a valuable tool that can be used to examine the pathophysiology of the disease and trial any potential treatment modalities. We present a methodology for the use of an experimental rat model that incorporates a guideline regarding radiation dose.

6.
J Craniomaxillofac Surg ; 48(7): 621-631, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32527622

ABSTRACT

PURPOSE: Osteoradionecrosis (ORN) is known to be a refractory disease in the oral and maxillofacial field. The purpose of this study was to examine the effects of pentoxifylline (PTX) and tocopherol (TP) on an ORN animal model focused on bone healing. MATERIALS AND METHODS: A total of 48 Sprague-Dawley rats were used: 40 received a single irradiation dose of 35 Gy on the left mandible, and eight were used as the nonirradiated control group. The rats received PTX (T1, C1), TP (T2, C2), a combination of PTX and TP (T3, C3), or normal saline (T4, C4). Three weeks after irradiation, the mandibular posterior teeth were extracted. The rats were sacrificed 4 weeks after extraction. RESULTS: In the T3 group, bone volume/tissue volume was 19.62 ± 16.03 (%), bone mineral density was as 0.31 ± 0.16 (g/cm3) in the micro-CT analysis, which were higher than that of other groups (p = 0.025, p = 0.012, respectively). In the histological analysis, bone regeneration was the most prominent in the T3 group. The ratio of empty lacunae was the highest in the T4 group, 68.77 ± 15.47 (%, p = 0.004). Immunohistochemistry showed that the expression of TNF-α was relatively lower in the T3 than in the T4 or T2 groups. The RT-qPCR showed the expression level of PECAM, VEGF-A, and osteocalcin was more than twofold as high as in the T3 group compared to the other groups. CONCLUSION: The combination of PTX and TP appears to promote angiogenesis and osteogenesis in a rat ORN model. Therefore, PTX and TP might be useful in the treatment and prevention of ORN.


Subject(s)
Osteoradionecrosis , Pentoxifylline , Animals , Disease Models, Animal , Rats , Rats, Sprague-Dawley , Tocopherols
7.
Arch Oral Biol ; 117: 104815, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32585446

ABSTRACT

AIM: To investigate the chemical and mechanical properties of teeth affected by a 1-bp deletion (c.2688delT) in the DSPP gene. METHODS AND MATERIALS: Maxillary first premolars were extracted from the affected individual at age 9 years due to the orthodontic reason for crowding. A sample was imbedded in epoxy resin and sectioned buccolingually, after micro-computerized tomography (µCT) images were taken. Scanning Electron Microscopy (SEM), Energy Dispersive Spectrometry (EDS) and Vickers microhardness testing were also performed. RESULTS: µCT reconstruction and analysis showed an irregularly obliterated pulp chamber and an extremely small pulpal volume in the DGI-II sample. The mineral density and microhardness scores were smaller in the dentin of the DGI-II sample compared to the wild-type. Mg content was lower in the dentin of the DGI-II sample compared to the wild-type. CONCLUSION: This study shows that dentin affected by a 1-bp deletion in DSPP has a reduced mineral density, diminished microhardness and reduced Mg content.


Subject(s)
Dentinogenesis Imperfecta , Extracellular Matrix Proteins/genetics , Phosphoproteins/genetics , Sialoglycoproteins/genetics , Child , Dentin/pathology , Dentinogenesis Imperfecta/diagnostic imaging , Dentinogenesis Imperfecta/genetics , Humans , Mutation , Pedigree , Sequence Deletion
8.
Proc Natl Acad Sci U S A ; 117(9): 4910-4920, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32071240

ABSTRACT

Growth and differentiation factor 11 (GDF11) and myostatin (MSTN) are closely related transforming growth factor ß (TGF-ß) family members, but their biological functions are quite distinct. While MSTN has been widely shown to inhibit muscle growth, GDF11 regulates skeletal patterning and organ development during embryogenesis. Postnatal functions of GDF11, however, remain less clear and controversial. Due to the perinatal lethality of Gdf11 null mice, previous studies used recombinant GDF11 protein to prove its postnatal function. However, recombinant GDF11 and MSTN proteins share nearly identical biochemical properties, and most GDF11-binding molecules have also been shown to bind MSTN, generating the possibility that the effects mediated by recombinant GDF11 protein actually reproduce the endogenous functions of MSTN. To clarify the endogenous functions of GDF11, here, we focus on genetic studies and show that Gdf11 null mice, despite significantly down-regulating Mstn expression, exhibit reduced bone mass through impaired osteoblast (OB) and chondrocyte (CH) maturations and increased osteoclastogenesis, while the opposite is observed in Mstn null mice that display enhanced bone mass. Mechanistically, Mstn deletion up-regulates Gdf11 expression, which activates bone morphogenetic protein (BMP) signaling pathway to enhance osteogenesis. Also, mice overexpressing follistatin (FST), a MSTN/GDF11 inhibitor, exhibit increased muscle mass accompanied by bone fractures, unlike Mstn null mice that display increased muscle mass without fractures, indicating that inhibition of GDF11 impairs bone strength. Together, our findings suggest that GDF11 promotes osteogenesis in contrast to MSTN, and these opposing roles of GDF11 and MSTN must be considered to avoid the detrimental effect of GDF11 inhibition when developing MSTN/GDF11 inhibitors for therapeutic purposes.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Bone and Bones/metabolism , Growth Differentiation Factors/metabolism , Muscle Development/physiology , Myostatin/metabolism , Osteogenesis/physiology , Animals , Bone Morphogenetic Proteins/genetics , Bone and Bones/pathology , Chondrocytes/metabolism , Down-Regulation , Follistatin , Gene Expression Regulation, Developmental , Growth Differentiation Factors/genetics , Mice , Mice, Knockout , Muscles/pathology , Osteoblasts/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
9.
Gene ; 733: 144274, 2020 Apr 05.
Article in English | MEDLINE | ID: mdl-31809844

ABSTRACT

Bone regeneration has been a challenge for both researchers and clinicians. In the field of tissue engineering, much effort has been made to identify cell sources including stem cells. The present study aimed to induce trans-differentiation from adipocytes to osteoblasts using epigenetic modifiers; 5-aza-dC and/or trichostatin-A (TSA). 3 T3-L1 preadipocytes were treated with TSA (100 nM) and then with Wnt3a (50 ng/ml). Microscopic observation showed trans-differentiated cell morphology. Methylation-specific PCR and immunoblotting were performed to analyze the DNA methylation and histone acetylation patterns. The gene expression was determined by real-time PCR. Based on these in vitro experiments, in vivo mouse experiments supplemented the possibility of trans-differentiation by epigenetic modification. TSA induced the acetylation of lysine9 on histone H3, and a sequential Wnt3a treatment stimulated the expression of bone marker genes in adipocytes, suppressing adipogenesis and stimulating osteogenesis. Furthermore, TSA induced DNA hypomethylation, and a combined treatment with TSA and 5-aza-dC showed a synergistic effect in epigenetic modifications. The number of adipocytes and DNA methylation patterns of old (15 months) and young (6 weeks) mice were significantly different, and TSA and sequential Wnt3a treatments increased bone formation in the old mice. Collectively, our results confirmed cell trans-differentiation via epigenetic modifications and osteogenic signaling from adipocytes to osteoblasts for the bone regeneration in vitro and in vivo, and indicated that histone acetylation could induce DNA hypomethylation, enhancing the chance of trans-differentiation.


Subject(s)
Adipocytes/metabolism , Cell Differentiation/drug effects , Osteoblasts/metabolism , 3T3-L1 Cells , Acetylation , Adipocytes/drug effects , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , CpG Islands , DNA Demethylation/drug effects , DNA Methylation/drug effects , DNA Methylation/genetics , Decitabine/metabolism , Decitabine/pharmacology , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Epigenomics/methods , Histone Deacetylase Inhibitors/pharmacology , Histones/genetics , Histones/metabolism , Hydroxamic Acids/metabolism , Hydroxamic Acids/pharmacology , Mice , Osteoblasts/drug effects , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics
10.
J Cell Physiol ; 234(12): 23360-23368, 2019 12.
Article in English | MEDLINE | ID: mdl-31183862

ABSTRACT

Growth and differentiation factor 11 (GDF11) is a transforming growth factor ß family member that has been identified as the central player of anterior-posterior (A-P) axial skeletal patterning. Mice homozygous for Gdf11 deletion exhibit severe anterior homeotic transformations of the vertebrae and craniofacial defects. During early embryogenesis, Gdf11 is expressed predominantly in the primitive streak and tail bud regions, where new mesodermal cells arise. On the basis of this expression pattern of Gdf11 and the phenotype of Gdf11 mutant mice, it has been suggested that GDF11 acts to specify positional identity along the A-P axis either by local changes in levels of signaling as development proceeds or by acting as a morphogen. To further investigate the mechanism of action of GDF11 in the vertebral specification, we used a Cdx2-Cre transgene to generate mosaic mice in which Gdf11 expression is removed in posterior regions including the tail bud, but not in anterior regions. The skeletal analysis revealed that these mosaic mice display patterning defects limited to posterior regions where Gdf11 expression is deficient, whereas displaying normal skeletal phenotype in anterior regions where Gdf11 is normally expressed. Specifically, the mosaic mice exhibited seven true ribs, a pattern observed in wild-type (wt) mice (vs. 10 true ribs in Gdf11-/- mice), in the anterior axis and nine lumbar vertebrae, a pattern observed in Gdf11 null mice (vs. six lumbar vertebrae in wt mice), in the posterior axis. Our findings suggest that GDF11, rather than globally acting as a morphogen secreted from the tail bud, locally regulates axial vertebral patterning.


Subject(s)
Body Patterning , Bone Morphogenetic Proteins/metabolism , Growth Differentiation Factors/metabolism , Osteogenesis , Spine/metabolism , Animals , Body Patterning/genetics , Bone Morphogenetic Proteins/deficiency , Bone Morphogenetic Proteins/genetics , Gene Expression Regulation, Developmental , Growth Differentiation Factors/deficiency , Growth Differentiation Factors/genetics , Mice, Inbred C57BL , Mice, Knockout , Mosaicism , Osteogenesis/genetics , Signal Transduction , Spine/embryology
11.
J Cell Biochem ; 120(2): 2226-2235, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30277585

ABSTRACT

Distal-less homeobox 3 (Dlx3), a member of the Dlx family of homeobox proteins, is a transcriptional activator of runt-related transcription factor 2 (Runx2) during osteogenic differentiation. It has been demonstrated that forced expression of Runx2 induces an osteogenic program and ectopic calcification in muscles. Therefore, it would be reasonable to predict that Dlx3 also affects myogenic differentiation. The relationship between Dlx3 and myogenesis, however, remains poorly understood. Therefore, in this study, the role and regulation of Dlx3 during myogenic differentiation were investigated. Expression level of Dlx3 was downregulated in human mesenchymal stem cells (MSCs), mouse MSCs, and C2C12 cells cultured in myogenic medium. Dlx3 level was inversely correlated with myogenic differentiation 1 and the muscle-specific microRNA, microRNA-133 (miR-133). The expression level of Runx2 was closely regulated by Dlx3 even under myogenic conditions. Overexpression of Dlx3 markedly downregulated expression levels of myogenic transcription factors and myotube formation in C2C12 cells, whereas Dlx3 knockdown enhanced myogenic differentiation. The Dlx3 3'-untranslated region (3'-UTR) has two potential binding sites for miR-133. Luciferase reporter assays demonstrated that Dlx3 is a direct target of miR-133a and miR-133b, and that the two target sites are redundantly active. Taken together, these results suggest that Dlx3 is a negative regulator of myogenic differentiation and that miR-133a and miR-133b enhance myogenic differentiation, partly through inhibition of Dlx3 expression via direct targeting of the Dlx3 3'-UTR.

12.
Adv Exp Med Biol ; 1064: 253-261, 2018.
Article in English | MEDLINE | ID: mdl-30471038

ABSTRACT

The fibrin matrix is fundamentally formed by the polymerization of fibrinogen and thrombin in blood plasma. It is a natural biopolymeric material to widely investigate for various tissue regenerations due to good biocompatibility, rapid biodegradability, and easy fabrication. In particular, the conjugated bioactive molecules with fibrinogen can promote tissue morphogenesis or maturation after cell adhesion on the matrices, migration, proliferation, or differentiation. Using these physiological properties with cell-material interactions, the fibrin matrices have been utilized in tissue engineering applications such as skin tissue, cardiovascular tissue, musculoskeletal tissue, or nerve tissue in preclinical and clinical situations. This chapter demonstrates the fibrin material and its tissue engineering applications as the therapeutic strategies.


Subject(s)
Biocompatible Materials/chemistry , Fibrin/chemistry , Regenerative Medicine , Tissue Engineering , Fibrinogen , Humans
13.
Int J Mol Sci ; 19(10)2018 Oct 14.
Article in English | MEDLINE | ID: mdl-30322210

ABSTRACT

Distal-less homeobox 5 (Dlx5) is a negative regulator of adipogenesis. Dlx5 expression is decreased by adipogenic stimuli, but the mechanisms of Dlx5 downregulation by adipogenic stimuli have not yet been determined. Here, we tested the impact of cAMP/PKA (protein kinase A) signaling induced by 3-isobutyl-1 methyl xanthine (IBMX), forskolin, and 8-CPT-cAMP on the expression of Dlx5 in 3T3-L1 preadipocytes. Significant downregulation of Dlx5 mRNA expression and protein production levels were observed via cAMP/PKA-dependent signaling. Forced expression of cAMP-responsive element-binding protein (CREB) and CCAAT/enhancer-binding protein ß (C/EBPß) was sufficient for downregulation of Dlx5 expression and revealed that CREB functions upstream of C/EBPß. In addition, C/EBPß knockdown by siRNA rescued Dlx5 expression in IBMX-treated 3T3-L1 preadipocytes. Luciferase assays using a Dlx5-luc-2935 reporter construct demonstrated the requirement of the Dlx5 promoter region, ranging from -774 to -95 bp that contains two putative C/EBPß binding elements (site-1: -517 to -510 bp and site-2: -164 to -157 bp), in the suppression of Dlx5 transcription. Consequently, chromatin immunoprecipitation analysis confirmed the importance of site-1, but not site-2, in C/EBPß binding and transcriptional suppression of Dlx5. In conclusion, we elucidated the underling mechanism of Dlx5 downregulation in IBMX-induced adipogenesis. IBMX activated cAMP/PKA/CREB signaling and subsequently upregulated C/EBPß, which binds to the Dlx5 promoter to suppress Dlx5 transcription.


Subject(s)
Adipocytes/cytology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Homeodomain Proteins/genetics , 1-Methyl-3-isobutylxanthine/pharmacology , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis , Animals , Binding Sites , CCAAT-Enhancer-Binding Protein-beta/genetics , Colforsin/pharmacology , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP Response Element-Binding Protein/genetics , Gene Expression Regulation/drug effects , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Mice , Signal Transduction/drug effects , Thionucleotides/pharmacology
14.
J Cell Physiol ; 233(12): 9390-9403, 2018 12.
Article in English | MEDLINE | ID: mdl-30132832

ABSTRACT

Myoblast fusion is critical for muscle growth, regeneration, and repair. We previously reported that the enzyme peptidyl-prolyl cis-trans isomerase NIMA interacting 1 (Pin1) is involved in osteoclast fusion. The objective of this study was to investigate the possibility that Pin1 also inhibits myoblast fusion. Here, we show the increased number of nuclei in the Pin1+/- mice muscle fiber compared to that in wild-type mice. Moreover, we show that low dose of the Pin1 inhibitor dipentamethylene thiuram monosulfide treatment caused enhanced fusion in C2C12 cells. The R-Smads are well-known mediators of muscle hypertrophy and hyperplasia as well as being substrates of Pin1. We found that Pin1 is crucial for maintaining the stability of Smad3 (homologues of the Drosophila protein, mothers against decapentaplegic (Mad) and the Caenorhabditis elegans protein Sma). Our results show that serine 204 within Smad3 is the key Pin1-binding site during inhibition of myoblast fusion and that both the transforming growth factor-ß receptor and extracellular signal-regulated kinase (ERK)-mediated phosphorylation are required for the interaction of Pin1 with Smad3. These findings suggest that a precise level of Pin1 activity is essential for regulating myoblast fusion during myogenesis and muscle regeneration.


Subject(s)
Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Smad3 Protein/metabolism , Animals , Cell Fusion , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Male , Mice, Inbred C57BL , Muscular Atrophy/genetics , Myoblasts/cytology , Myoblasts/metabolism , Myostatin/metabolism , Phosphorylation , Protein Binding , Serine/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
15.
ACS Appl Mater Interfaces ; 10(21): 17526-17541, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29741358

ABSTRACT

Nanofibrous engineered matrices have significant potential in cellular differentiation and tissue regeneration. Stem cells require specific extracellular signals that lead to the induction of different lineages. However, the mechanisms by which the nanofibrous matrix promotes mesenchymal stem cell (MSC) differentiation are largely unknown. Here, we investigated the mechanisms that underlie nanofibrous matrix-induced odontoblastic differentiation of human dental pulp MSCs (DP-MSCs). An electrospun polystyrene nanofibrous (PSF) matrix was prepared, and the cell responses to the PSF matrix were assessed in comparison with those on conventional tissue culture dishes. The PSF matrix promoted the expression of Wnt3a, Wnt5a, Wnt10a, BMP2, BMP4, and BMP7 in the DP-MSCs, concomitant with the induction of odontoblast/osteoblast differentiation markers, dentin sialophosphoprotein (DSPP), osteocalcin, and bone sialoprotein, whose levels were further enhanced by treatment with recombinant Wnt3a. The DP-MSCs cultured on the PSF matrix also exhibited a high alkaline phosphatase activity and intense Alizarin Red staining, indicating that the PSF matrix promotes odontoblast differentiation. Besides inducing the expression of Wnt3a, the PSF matrix maintained high levels of ß-catenin protein and enhanced its translocation to the nucleus, leading to its transcriptional activity. Forced expression of LEF1 or treatments with LiCl further enhanced the DSPP expression. Blocking the Wnt3a-initiated signaling abrogated the PSF-induced DSPP expression. Furthermore, the cells on the PSF matrix increased the DSPP promoter activity. The ß-catenin complex was bound to the conserved motifs on the DSPP promoter dictating its transcription. Transplantations of the preodontoblast-seeded PSF matrix to the subcutaneous tissues of nude mice confirmed the association of the PSF matrix with the Wnt3a and DSPP expressions in vivo. Taken together, these results demonstrate the nanofibrous engineered matrix strongly supports odontoblastic differentiation of DP-MSCs by enhancing Wnt/ß-catenin signaling.


Subject(s)
Stem Cells , Animals , Cell Differentiation , Dental Pulp , Extracellular Matrix Proteins , Humans , Mice , Mice, Nude , Wnt Signaling Pathway
16.
Int J Mol Sci ; 19(1)2018 Jan 09.
Article in English | MEDLINE | ID: mdl-29315243

ABSTRACT

Hyperglycemic conditions in diabetic patients can affect various cellular functions, including the modulation of osteogenic differentiation. However, the molecular mechanisms by which hyperglycemia affects osteogenic differentiation are yet to be clarified. This study aimed to investigate whether the aberrant increase in protein O-linked-ß-N-acetylglucosamine glycosylation (O-GlcNAcylation) contributes to the suppression of osteogenic differentiation due to hyperglycemia. To induce osteogenic differentiation, C2C12 cells were cultured in the presence of recombinant human bone morphogenetic protein 2 (BMP2). Excessive protein O-GlcNAcylation was induced by treating C2C12 cells with high glucose, glucosamine, or N-acetylglucosamine concentrations or by O-GlcNAc transferase (OGT) overexpression. The effect of O-GlcNAcylation on osteoblast differentiation was then confirmed by examining the expression levels of osteogenic marker gene mRNAs, activity of alkaline phosphatase, and transcriptional activity of Runx2, a critical transcription factor for osteoblast differentiation and bone formation. Cell treatment with high glucose, glucosamine or N-acetylglucosamine increased O-GlcNAcylation of Runx2 and the total levels of O-GlcNAcylated proteins, which led to a decrease in the transcriptional activity of Runx2, expression levels of osteogenic marker genes (Runx2, osterix, alkaline phosphatase, and type I collagen), and activity of alkaline phosphatase. These inhibitory effects were rescued by lowering protein O-GlcNAcylation levels by adding STO45849, an OGT inhibitor, or by overexpressing ß-N-acetylglucosaminidase. Our findings suggest that excessive protein O-GlcNAcylation contributes to high glucose-suppressed osteogenic differentiation.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Cell Differentiation/drug effects , Osteogenesis/drug effects , Transforming Growth Factor beta/pharmacology , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Enzyme Inhibitors/pharmacology , Glucosamine/pharmacology , Glucose/pharmacology , Glycosylation/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Periodontal Ligament/metabolism , Recombinant Proteins/pharmacology , Sp7 Transcription Factor/genetics , Sp7 Transcription Factor/metabolism , Transcription, Genetic/drug effects
17.
J Endod ; 44(1): 98-103.e1, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29246377

ABSTRACT

INTRODUCTION: The in vivo effect of prolyl hydroxylase inhibitors on the regeneration of the pulp-dentin complex is unclear. The purpose of this study was to investigate the effect of dimethyloxalylglycine (DMOG)-embedded poly(ε-caprolactone) fiber (PCLF/DMOG) on odontoblastic differentiation of human dental pulp-derived cells (hDPCs) by transplantation of the dentin slice model. METHODS: The hDPCs were seeded onto electrospun PCLF and PCLF/DMOG in dentin slices and then transplanted into nude mice. The surface topography was evaluated for both PCLFs, and DMOG release from the PCLF/DMOG was examined. The effects of the PCLF/DMOG were assessed by histology and quantitative reverse transcription polymerase chain reaction. RESULTS: The PCLF/DMOG-treated dentin slices showed higher cellularity with a palisading arrangement of hDPCs and organized collagen fibers. We found that the PCLF/DMOG significantly stimulated the expression of vascular endothelial growth factor, dentin sialoprotein, and bone sialoprotein in the hDPCs (P < .05) and mouse vascular endothelial growth factor A, mouse platelet endothelial cell adhesion molecule 1, and mouse neurofilament light polypeptide in the surrounding host cells (P < .05). CONCLUSIONS: These results show that PCLF/DMOG has potential in pulp-dentin complex regeneration by promoting odontoblastic differentiation of hDPCs and by enhancing host cell recruitment, angiogenesis, and neurogenesis through the released DMOG-mediated cell responses.


Subject(s)
Amino Acids, Dicarboxylic , Dental Pulp/cytology , Odontoblasts/cytology , Polyesters , Animals , Cell Differentiation , Cells, Cultured , Humans , Mice , Mice, Nude , Surgical Mesh
18.
J Med Food ; 21(1): 57-69, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28981378

ABSTRACT

Morinda citrifolia (Noni) leaf is an herbal medicine with application in the domestic treatment of a broad range of conditions, including bone fracture and luxation. However, the basic mechanism underlying the stimulation of osteogenic differentiation by Noni leaf extract remains poorly understood. This study aimed to examine the effect of this extract on osteogenic differentiation and the mechanism by which Noni leaf extract enhances osteogenic differentiation. Aqueous extract of Noni leaves was prepared, and rutin and kaempferol-3-O-rutinoside were identified to be two of its major components. C2C12 and human periodontal ligament (hPDL) cells were used to study the effect of Noni. Noni did not show cytotoxicity at a concentration range of 0.015%-1.0% (w/v%) and significantly enhanced the activity of alkaline phosphatase (ALP) and expression levels of osteoblast differentiation markers, including Runx2, ALP, osterix, and osteocalcin, bone morphogenetic protein 2, Wnt3a, and ß-catenin. In addition, Noni enhanced the matrix mineralization of hPDL cells. In the signaling pathways, Noni increased the phosphorylation levels of Akt and GSK3ß and nuclear translocation and transcriptional activity of ß-catenin, which were attenuated by the addition of Dkk-1, a Wnt inhibitor, or LY294002, a PI3K inhibitor. These results suggest that Noni leaf extract enhances osteogenic differentiation through the PI3K/Akt-dependent activation of Wnt/ß-catenin signaling. Noni leaf extract might be a novel alternative medicine for bone and periodontal regeneration in patients with periodontal diseases.


Subject(s)
Morinda/chemistry , Osteoblasts/drug effects , Osteogenesis/drug effects , Plant Extracts/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cell Line , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocalcin/genetics , Osteocalcin/metabolism , Plant Leaves/chemistry , Wnt3A Protein/genetics , Wnt3A Protein/metabolism , beta Catenin/genetics
19.
Int J Mol Sci ; 18(11)2017 Nov 09.
Article in English | MEDLINE | ID: mdl-29120400

ABSTRACT

Cementum is a mineralized layer on the tooth's root surface and facilitates the biomechanical anchoring of fibrous connective tissues as a part of tooth-supportive complexes. Previously, we observed that OCCM30 cementoblasts cultured on fibrin matrices underwent apoptosis due to fibrin degradation through the expression of proteases. Here, we demonstrated that OCCM30 on fibrin matrices (OCCM30-fibrin) enhanced canonical Wnt signaling, which directed to plasminogen expression. The OCCM30-fibrin showed higher levels of Wnt3a expression, nuclear translocation of ß-catenin, and T-cell factor (TCF) optimal motif (TOP) reporter activity than the cells on tissue culture dishes (OCCM30-TCD), indicating that the OCCM30-fibrin enhanced canonical Wnt/ß-catenin signaling. Also, OCCM30-fibrin expressed biomineralization-associated markers at higher levels than OCCM30-TCD, of which levels were further increased with LiCl, a Wnt signaling activator. The OCCM30 cementoblasts simultaneously showed that high levels of plasminogen, a critical component of fibrinolysis, were expressed in the OCCM30-fibrin. Activation of canonical Wnt signaling with LiCl treatment or with forced lymphoid enhancer factor 1 (LEF1)-expression increased the expression of plasminogen. On the contrary, the inhibition of canonical Wnt signaling with siRNAs against Wnt3a or ß-catenin abrogated fibrin-enhanced plasminogen expression. Furthermore, there are three conserved putative response elements for the LEF1/ß-catenin complex in the plasminogen proximal promoter regions (-900 to +54). Site-directed mutations and chromatin immunoprecipitation indicated that canonical Wnt signaling directed plasminogen expression. Taken together, this study suggests that fibrin-based materials can modulate functional periodontal formations in controlling cementoblast differentiation and fibrin degradation.


Subject(s)
Dental Cementum/metabolism , Fibrin/metabolism , Plasminogen/metabolism , Wnt Signaling Pathway , Animals , Biomarkers/analysis , Cell Line , Fibrin/genetics , Fibrinolysis/drug effects , Lithium Chloride/pharmacology , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Plasminogen/genetics , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , beta Catenin/genetics , beta Catenin/metabolism
20.
Biomaterials ; 149: 77-87, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29017079

ABSTRACT

Host responses to a biomaterial critically influence its in vivo performance. Biomaterial architectures that can recruit endogenous host stem cells could be beneficial in tissue regeneration or integration. Here, we report that the fibrous topography of biomaterials promotes the recruitment of host mesenchymal stem cells (MSCs) by facilitating the macrophage phenotype transition from M1-to-M2. Electrospun poly (ε-caprolactone) fiber (PCL-fiber) films were implanted into the subcutaneous tissues of rats, and the response of host cells to the PCL-fiber was evaluated and compared with those of solid ones (PCL-solid). During the initial post-implantation period, greater numbers of cells were recruited and adhered to the PCL-fiber compared to the PCL-solid, and the cells exhibited the M1 phenotype, which was supported by the enhanced adsorption of complement C3a to the implanted PCL-fiber. Subsequently, the PCL-fiber supported the macrophage phenotype transition from M1-to-M2, which was confirmed by the ratio of M2/M1 marker (CD163/CCR7)-positive cells and by the expression of M2/M1 markers (arginase-1/iNOS). The PCL-fiber also reduced the formation of foreign body giant cells. MSC marker (CD29, CD44, and CD90)-positive cells began to appear as early as day 4 on the PCL-fiber, while few MSCs were observed on the PCL-solid. The MSCs migration ex vivo assay showed that MSCs substantially migrated across the trans-wells toward the implanted PCL-fiber. The cells on the implanted PCL-fiber expressed and secreted substantial levels of SDF-1 (CXCL-12), while anti-SDF-1 neutralizing antibody abrogated the MSCs migration. Taken together, these results provide evidence that the fibrous topography of biomaterials enhances the recruitment of MSCs by promoting macrophage recruitment, facilitating M1-to-M2 transition, and enhancing SDF-1 secretion.


Subject(s)
Macrophages/cytology , Mesenchymal Stem Cells/physiology , Polyesters/chemistry , Animals , Biomarkers/metabolism , Cell Adhesion , Cell Movement , Chemokine CXCL12/metabolism , Humans , Macrophages/physiology , Male , Mesenchymal Stem Cells/cytology , Phenotype , Rats, Sprague-Dawley , Tissue Scaffolds
SELECTION OF CITATIONS
SEARCH DETAIL
...