Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Mol Cell Cardiol ; 190: 48-61, 2024 May.
Article in English | MEDLINE | ID: mdl-38582260

ABSTRACT

We have demonstrated that directly reprogramming cardiac fibroblasts into new cardiomyocytes via miR combo improves cardiac function in the infarcted heart. However, major challenges exist with delivery and efficacy. During a screening based approach to improve delivery, we discovered that C166-derived EVs were effective delivery agents for miR combo both in vitro and in vivo. In the latter, EV mediated delivery of miR combo induced significant conversion of cardiac fibroblasts into cardiomyocytes (∼20%), reduced fibrosis and improved cardiac function in a myocardial infarction injury model. When compared to lipid-based transfection, C166 EV mediated delivery of miR combo enhanced reprogramming efficacy. Improved reprogramming efficacy was found to result from a miRNA within the exosome: miR-148a-3p. The target of miR-148a-3p was identified as Mdfic. Over-expression and targeted knockdown studies demonstrated that Mdfic was a repressor of cardiomyocyte specific gene expression. In conclusion, we have demonstrated that C166-derived EVs are an effective method for delivering reprogramming factors to cardiac fibroblasts and we have identified a novel miRNA contained within C166-derived EVs which enhances reprogramming efficacy.


Subject(s)
Cellular Reprogramming , Fibroblasts , MicroRNAs , Myocytes, Cardiac , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Cellular Reprogramming/genetics , Myocytes, Cardiac/metabolism , Fibroblasts/metabolism , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Exosomes/metabolism , Gene Expression Regulation , Humans
2.
Front Bioeng Biotechnol ; 12: 1370234, 2024.
Article in English | MEDLINE | ID: mdl-38456010

ABSTRACT

Insufficient physical activity poses a significant risk factor for cardiovascular diseases. Exercise plays a crucial role in influencing the vascular system and is essential for maintaining vascular health. Hemodynamic stimuli generated by exercise, such as shear stress and circumferential stress, directly impact vascular structure and function, resulting in adaptive changes. In clinical settings, incorporating appropriate exercise interventions has become a powerful supplementary approach for treating and rehabilitating various cardiovascular conditions. However, existing models for studying exercise-induced vascular adaptation primarily rely on in vivo animal and in vitro cellular models, each with its inherent limitations. In contrast, human research faces challenges in conducting mechanistic analyses due to ethics issues. Therefore, it is imperative to develop highly biomimetic in vitro/ex vivo vascular models that can replicate exercise stimuli in human systems. Utilizing various vascular assessment techniques is also crucial to comprehensively evaluate the effects of exercise on the vasculature and uncover the molecular mechanisms that promote vascular health. This article reviews the hemodynamic mechanisms that underlie exercise-induced vascular adaptation. It explores the advancements in current vascular models and measurement techniques, while addressing their future development and challenges. The overarching goal is to unravel the molecular mechanisms that drive the positive effects of exercise on the cardiovascular system. By providing a scientific rationale and offering novel perspectives, the aim is to contribute to the formulation of precise cardiovascular rehabilitation exercise prescriptions.

3.
Front Endocrinol (Lausanne) ; 14: 1219194, 2023.
Article in English | MEDLINE | ID: mdl-37501791

ABSTRACT

Maternal exercise during pregnancy has emerged as a potentially promising approach to protect offspring from cardiovascular disease, including hypertension. Although endothelial dysfunction is involved in the pathophysiology of hypertension, limited studies have characterized how maternal exercise influences endothelial function of hypertensive offspring. In this study, pregnant spontaneously hypertensive rats and Wistar-Kyoto rats were assigned either to a sedentary lifestyle or to swimming training daily, and fetal histone deacetylase-mediated epigenetic modification and offspring vascular function of mesenteric arteries were analyzed. Maternal exercise ameliorated the impairment of acetylcholine-induced vasodilation without affecting sodium nitroprusside-induced vasodilation in mesenteric arteries from the hypertensive offspring. In accordance, maternal exercise reduced NADPH oxidase-4 (Nox4) protein to prevent the loss of nitric oxide generation and increased reactive oxygen species production in mesenteric arteries of hypertensive offspring. We further found that maternal exercise during pregnancy upregulated vascular SIRT1 (sirtuin 1) expression, leading to a low level of H3K9ac (histone H3 lysine 9 acetylation), resulting in the transcriptional downregulation of Nox4 in mesenteric arteries of hypertensive fetuses. These findings show that maternal exercise alleviates oxidative stress and the impairment of endothelium-dependent vasodilatation via SIRT1-regulated deacetylation of Nox4, which might contribute to improved vascular function in hypertensive offspring.


Subject(s)
Hypertension , Hypotension , NADPH Oxidase 4 , Physical Conditioning, Animal , Sirtuin 1 , Animals , Female , Pregnancy , Rats , Endothelium, Vascular , Hypertension/prevention & control , NADPH Oxidase 4/genetics , Oxidative Stress , Rats, Inbred SHR , Rats, Inbred WKY , Sirtuin 1/genetics
4.
J Biol Chem ; 299(5): 104694, 2023 05.
Article in English | MEDLINE | ID: mdl-37044217

ABSTRACT

Directly reprogramming fibroblasts into cardiomyocytes improves cardiac function in the infarcted heart. However, the low efficacy of this approach hinders clinical applications. Unlike the adult mammalian heart, the neonatal heart has an intrinsic regenerative capacity. Consequently, we hypothesized that birth imposes fundamental changes in cardiac fibroblasts which limit their regenerative capabilities. In support, we found that reprogramming efficacy in vitro was markedly lower with fibroblasts derived from adult mice versus those derived from neonatal mice. Notably, fibroblasts derived from adult mice expressed significantly higher levels of pro-angiogenic genes. Moreover, under conditions that promote angiogenesis, only fibroblasts derived from adult mice differentiated into tube-like structures. Targeted knockdown screening studies suggested a possible role for the transcription factor Epas1. Epas1 expression was higher in fibroblasts derived from adult mice, and Epas1 knockdown improved reprogramming efficacy in cultured adult cardiac fibroblasts. Promoter activity assays indicated that Epas1 functions as both a transcription repressor and an activator, inhibiting cardiomyocyte genes while activating angiogenic genes. Finally, the addition of an Epas1 targeting siRNA to the reprogramming cocktail markedly improved reprogramming efficacy in vivo with both the number of reprogramming events and cardiac function being markedly improved. Collectively, our results highlight differences between neonatal and adult cardiac fibroblasts and the dual transcriptional activities of Epas1 related to reprogramming efficacy.


Subject(s)
Cellular Reprogramming , Myocytes, Cardiac , Transcription Factors , Animals , Mice , Fibroblasts/cytology , Gene Expression Regulation , Myocytes, Cardiac/cytology , Transcription Factors/metabolism , Animals, Newborn
5.
Bone ; 153: 116162, 2021 12.
Article in English | MEDLINE | ID: mdl-34455116

ABSTRACT

DNA damage-inducible transcript 3 (DDIT3), a member of the CCAAT/enhancer-binding protein (C/EBP) family, is involved in cellular apoptosis and differentiation. DDIT3 participates in the regulation of adipogenesis and osteogenesis in vitro and in vivo. However, the role of DDIT3 in osteoclastogenesis is not yet known. In this study, the involvement of DDIT3 in osteoclast differentiation and function was reported for the first time. CRISPR/Cas9-mediated DDIT3 knockout (KO) mice were generated for functional assessment. Tartrate-resistant acid phosphatase (TRAP) staining of distal femurs showed increased positive cells in DDIT3 KO mice. DDIT3 expression was downregulated during the receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclast differentiation of bone marrow-derived macrophages (BMMs). The loss of DDIT3 increased the expression of osteoclast-specific markers, including nuclear factor of activated T-cells cytoplasmic 1 (NFATc1), TRAP, cathepsin K (CTSK), and dendritic cell-specific transmembrane protein (DC-STAMP) and promoted the formation of TRAP-positive multinucleated osteoclasts. The actin ring number and resorption area of bone slices were also increased in DDIT3 KO BMMs. Lentivirus-mediated DDIT3 overexpression significantly inhibited the osteoclast differentiation of RAW264.7 cells. In the tumor necrosis factor-α-induced osteolysis model, DDIT3 deficiency enhanced osteoclast formation and aggravated bone resorption. DDIT3 inhibited osteoclast differentiation by regulating the C/EBPα-CTSK axis. Furthermore, DDIT3 KO intensified the RANKL-triggered activation of the MAPKs and Akt signaling pathways. Taken together, the results revealed the essential role of DDIT3 in osteoclastogenesis in vitro and in vivo and its close relationship with osteoclast-associated transcription factors and pathways.


Subject(s)
Bone Resorption , Osteolysis , Animals , Bone Resorption/genetics , Cell Differentiation , DNA Damage , Mice , NFATC Transcription Factors , Osteoclasts , Osteogenesis , RANK Ligand
7.
Am J Physiol Cell Physiol ; 319(6): C1141-C1150, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33026832

ABSTRACT

Stem cell injections are an attractive therapeutic tool. It has been demonstrated that injected stem cells promote tissue repair and regeneration via paracrine mechanisms. However, the effects of injected stem cells continue for far longer than they are present. We hypothesized that the effects of injected stem cells are prolonged because of a sequential paracrine relay mechanism. Conditioned media was collected from mesenchymal stem cells (MSCs) after 24 h. This media was then added to RAW264.7. Media was collected from the macrophages after 24 h and was then added to endothelial cells (ECs). This conditioned macrophage media, but not control media, promoted wound healing and induced EC differentiation. Similar results were observed with primary macrophages. To identify the active paracrine factors released by macrophages in response to stimulation by MSC conditioned media we used an antibody array, identifying increased expression of the angiogenesis-related proteins stromal cell-derived factor 1 (SDF1) and plasminogen activator inhibitor-1 (PAI-1). Knockdown of either protein inhibited the ability of conditioned media derived from MSC paracrine factor-stimulated macrophages to induce EC differentiation both in vitro and in vivo. Conditioned media derived from postnatal day 7 (P7) mouse macrophages induced EC differentiation. Moreover, SDF1 and PAI-1 levels were >120 higher in P7 macrophages compared with adult macrophages, suggesting that MSC paracrine factors promote adult macrophages to adopt a juvenile phenotype. These results indicate that MSC paracrine factors induce macrophages to secrete SDF1 and PAI-1, in-turn inducing endothelial cells to differentiate. Identification of a sequential paracrine mechanism opens new therapeutic avenues for stem cell therapy.


Subject(s)
Culture Media, Conditioned/pharmacology , Endothelial Cells/drug effects , Guided Tissue Regeneration/methods , Mesenchymal Stem Cell Transplantation , Paracrine Communication/physiology , Wound Healing/drug effects , Animals , Cell Differentiation/drug effects , Cell- and Tissue-Based Therapy/methods , Chemokine CXCL12/metabolism , Macrophages/cytology , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , RAW 264.7 Cells , Serpin E2/metabolism
8.
J Cell Mol Med ; 24(14): 7939-7948, 2020 07.
Article in English | MEDLINE | ID: mdl-32510818

ABSTRACT

Cementum regeneration, as one of the most difficult challenges of periodontal regeneration, is influenced by inflammatory factors. Inflammation may hamper or promote periodontal tissue repair under different circumstances, as it is found to do in dentin-pulp complex and bone tissue. Our team demonstrated that YAP promotes mineralization of OCCM, a cementoblast cell line. However, the effect of YAP on its mineralization under inflammatory microenvironment is unclear. In this study, cementogenesis in vitro was up-regulated after transient TNF-α treatment for 30 minutes. YAP expression also was increased by TNF-α treatment. YAP overexpression promoted OCCM mineralization after the cells were transiently treated with TNF-α because YAP overexpression inhibited NF-κB pathway activity, while YAP knockdown elevated it. The inhibited mineralization potential and activated NF-κB pathway activity by YAP knockdown also were partly rescued by the application of the NF-κB inhibitor Bay 11-7082. These results demonstrated that YAP plays a positive role in the mineralization of TNF-α transiently treated cementoblast, partly by inhibiting the NF-κB pathway activity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcification, Physiologic/drug effects , Cell Cycle Proteins/metabolism , Cementogenesis , NF-kappa B/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins/genetics , Cell Line , Cementogenesis/drug effects , Cytokines/metabolism , Fluorescent Antibody Technique , Gene Expression , Gene Knockdown Techniques , Inflammation Mediators/metabolism , Mice , YAP-Signaling Proteins
9.
ACS Appl Bio Mater ; 3(6): 3569-3581, 2020 Jun 15.
Article in English | MEDLINE | ID: mdl-35025227

ABSTRACT

The regeneration of craniofacial bone defects remains a crucial clinical challenge. To date, numerous biomaterials are applied in this field. However, current strategies have ignored the importance of intramembranous ossification and the vital role of macrophages in regulating osteogenesis. Here, an osteoblast (OB)-targeting peptide (SDSSD)-modified chitosan scaffold (CS-SDSSD) is developed for imitating the physiological process of bone development from the fibrous membrane. The addition of free peptide (fSDSSD) can recruit host OBs, and the peptide grafted on the scaffold (CS-SDSSD) can well organize the migrated OBs by binding with their surface periostin. Besides, macrophage polarization is found in the bone defects. CS-SDSSD + fSDSSD displays advantages in prioritizing M2 macrophage polarization and promoting the intramembranous ossification bone repair process. In summary, our strategy provides an economical and effective path for craniofacial bone repair and holds great potential for biomedical applications.

10.
J Cell Physiol ; 235(5): 4698-4708, 2020 05.
Article in English | MEDLINE | ID: mdl-31642068

ABSTRACT

Yes-associated protein 1 (YAP1), the core downstream effector of the Hippo signaling cascade, was involved in the regulation of osteoblast and osteoclast differentiation and in bone metabolism. However, the regulatory effects and mechanisms of YAP1 on bone-remodeling molecules in osteoblasts under inflammation remain unknown. In this study, YAP1 expression level was downregulated after treatment with inflammatory cytokine tumor necrosis factor-α (TNF-α) in MC3T3-E1 cells. The key osteoclastogenic molecules induced by TNF-α, namely, interleukin-6 and receptor activator of nuclear factor-κB (NF-κB) ligand, were suppressed after lentivirus-induced YAP1 overexpression, which dramatically increased the expression level of osteoprotegerin. Conversely, the expression levels of the above factors showed opposite trends in the YAP1 small interfering RNA and YAP1 inhibitor (verteporfin) group. Mechanistically, YAP1 attenuated the TNF-α-induced activation of the NF-κB signaling pathway as revealed by the reduced expression of phosphorylated-p65 and NF-κB reporter activity and the nuclear translocation of p65. Moreover, the expression level of YAP1 suppressed by TNF-α was reversed by berberine in concentration-dependent manner. Taken together, our study suggests that YAP1 plays a critical role in the regulation of bone metabolism and is a potential therapeutic target for treating inflammatory bone resorption.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone Remodeling/drug effects , Bone Resorption/metabolism , Cell Cycle Proteins/metabolism , NF-kappa B/metabolism , Osteoblasts/drug effects , Tumor Necrosis Factor-alpha/pharmacology , 3T3 Cells , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Resorption/genetics , Bone Resorption/physiopathology , Cell Cycle Proteins/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , NF-kappa B/genetics , Osteoblasts/metabolism , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , Signal Transduction , YAP-Signaling Proteins
11.
J Cell Physiol ; 234(12): 22719-22730, 2019 12.
Article in English | MEDLINE | ID: mdl-31131439

ABSTRACT

Tooth cementum is a bone-like mineralized tissue and serves as a microbial barrier against invasion and destruction. Cementum is also responsible for tooth stability and defending pulp from outside stimuli, which is formed by cementoblasts. Although it is crucial for periodontal and periapical diseases, the mechanisms underlying the pathophysiological changes of cementoblasts and their inflammatory responses remain unclear. MiR-181b is found to modulate vascular inflammation and endotoxin tolerance. In this study, miR-181b-5p was downregulated in tumor necrosis factor-α (TNF-α)-stimulated cementoblasts, whereas proinflammatory molecules increased. The mouse periapical lesions have similar results, which imitate an inflammatory environment for cementoblasts in vivo. The bioinformatics analysis and dual luciferase reporter assay suggested that miR-181b-5p targeted interleukin-6 (IL-6). Overexpressing miR-181b-5p negatively regulated IL-6 and proinflammatory chemokine. Western blot analysis and luciferase activity reporter assay verified that miR-181b-5p weakened the NF-κB activity. Hence, miR-181b-5p moderated proinflammatory chemokine production by targeting IL-6 in cementoblasts and NF-κB signaling pathway was involved. Furthermore, miR-181b-5p promoted cementoblast apoptosis, which may enhance the resolution of inflammation. Overall, our data revealed that miR-181b-5p was a negative regulator of TNF-α-induced inflammatory responses in cementoblasts.


Subject(s)
Dental Cementum/drug effects , Interleukin-6/metabolism , MicroRNAs/metabolism , Periodontitis/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Dental Cementum/immunology , Dental Cementum/metabolism , Dental Cementum/pathology , Disease Models, Animal , Gene Expression Regulation , Interleukin-6/genetics , Mice , MicroRNAs/genetics , NF-kappa B/metabolism , Periodontitis/genetics , Periodontitis/immunology , Periodontitis/pathology , Signal Transduction
12.
Am J Transl Res ; 11(1): 314-326, 2019.
Article in English | MEDLINE | ID: mdl-30787989

ABSTRACT

Angiopoietin-like protein 2 (ANGPTL2) is abundantly expressed in adipose tissue, is associated with tissue homeostasis, and promotes osteoblast and chondrocyte differentiation. In teeth, cementum, a thin layer of mineralized tissue that is formed by cementoblasts, covers the entire root surface and is a vital component of periodontium. The cementoblasts regulate the deposition and mineralization of the cementum matrix. However, the effects of ANGPTL2 on cementoblast differentiation have not been studied. The objective of this study was to elucidate the role of ANGPTL2 during cementoblast differentiation and determine its underlying mechanisms. Our results showed that the expression levels of ANGPTL2 gradually increased during cementoblast differentiation. After ANGPTL2 was knocked down using short-hairpin RNA, the levels of the osteogenic markers osterix (OSX), alkaline phosphatase (ALP), bone sialoprotein (BSP), and osteocalcin (OCN) decreased. In addition, ALP activity and the number of calcified nodules were dramatically reduced compared with those in the negative control. Interestingly, the ERK1/2 signaling pathway was activated after ANGPTL2 knockdown. Treatment with PD98059, the inhibitor of the ERK1/2 signaling pathway, partially rescued the decreased differentiation capability of cementoblast caused by ANGPTL2 downregulation. Collectively, ANGPTL2 knockdown inhibited cementoblast differentiation partially by activating the ERK1/2 signaling pathway. These findings suggest that ANGPTL2 was indispensable in cementoblast differentiation.

13.
J Cell Physiol ; 234(8): 14007-14018, 2019 08.
Article in English | MEDLINE | ID: mdl-30618072

ABSTRACT

Yes-associated protein 1 (YAP1) transcriptional coactivator has recently been identified to regulate skeletal lineage cell differentiation and bone development. However, the role and molecular mechanisms of YAP1 in the regulation of osteoblastic differentiation remains to be elucidated. In this study, we demonstrated that YAP1 expression was increased during osteogenic differentiation of rat bone mesenchymal stem cells and MC3T3-E1. YAP1 overexpression MC3T3-E1 showed increased expression of osteogenesis markers, such as runt-related transcription factor 2, osteocalcin, and osteopontin, as well as alkaline phosphatase and alizarin red staining. Conversely, YAP1 knockdown significantly suppressed MC3T3-E1 osteoblastic differentiation. Mechanistically, we found that YAP1 overexpression upregulated the mRNA and protein expression of the inhibitor of differentiation/DNA binding 1 (ID1), which was contrary to the results of YAP1-knockdown group. Moreover, the early osteogenic differentiation of MC3T3-E1 cells was enhanced by ID1 overexpression. Furthermore, transient transfection with exogenous ID1 overexpression plasmid completely recaptured the decreased effects of YAP1 knockdown on MC3T3-E1 cell differentiation. In addition, ß-catenin and AMP-activated protein kinase signaling pathways participated in YAP1 regulation processes. Taken together, our study suggests that YAP1 is a crucial modulator of osteoblast differentiation in vitro, and provides insight into the mechanism by which YAP1 regulates osteoblast differentiation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Cell Differentiation , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Adenylate Kinase/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Cell Proliferation , Down-Regulation/genetics , Inhibitor of Differentiation Protein 1/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Models, Biological , Osteogenesis/genetics , Rats, Sprague-Dawley , Signal Transduction , Transcription, Genetic , Up-Regulation/genetics , YAP-Signaling Proteins , beta Catenin/metabolism
14.
J Cell Physiol ; 234(7): 11602-11609, 2019 07.
Article in English | MEDLINE | ID: mdl-30488444

ABSTRACT

DDIT3 is of great importance in endoplasmic reticulum stress and is involved in many inflammatory diseases and mineralization processes. The cementum protects teeth from periodontitis and provides attachment for Sharpey's fibers of the periodontal ligament. However, the effect of DDIT3 on cementoblast differentiation remains largely unknown. In this study, we found that DDIT3 was suppressed during cementoblast differentiation. Knockdown of DDIT3 increased the messenger RNA (mRNA) and protein levels of several key osteogenic markers in vitro, including alkaline phosphatase, runt-related transcription factor 2, and osteocalcin (OCN). In addition, isocitrate dehydrogenase 1 (IDH1) was increased during cementoblast differentiation, and knockdown of DDIT3 increased the protein and mRNA levels of IDH1. Furthermore, inhibition of IDH1 could partially reduce the effect of DDIT3 on cementoblast differentiation. The DDIT3 knockdown activated nuclear factor-κB (NF-κB) transcriptional activity and upregulated the expression of p-p65 and p-IκBα. The increased osteogenic differentiation ability and IDH1 expression, as induced by the DDIT3 knockdown, could be partially turned over by the addition of NF-κB inhibitor BAY 11-7082. Overall, our data clarified that DDIT3 suppresses cementoblast differentiation through IDH1, via the NF-κB pathway.


Subject(s)
Dental Cementum/metabolism , Isocitrate Dehydrogenase/metabolism , NF-kappa B/metabolism , Transcription Factor CHOP/metabolism , Animals , Cell Differentiation , Cell Line , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Isocitrate Dehydrogenase/genetics , Mice , NF-kappa B/genetics , Nitriles/pharmacology , Sulfones/pharmacology , Transcription Factor CHOP/genetics
15.
J Cell Physiol ; 234(4): 4005-4014, 2019 04.
Article in English | MEDLINE | ID: mdl-30256390

ABSTRACT

Cementum, which shares common features with bone in terms of biochemical composition, is important for the homeostasis of periodontium during periodontitis and orthodontic treatment. Sirtuin 6 (SIRT6), as a member of the sirtuin family, plays key roles in the osteogenic differentiation of bone marrow mesenchymal stem cells. However, the involvement of SIRT6 in cementoblast differentiation and mineralization and the underlying mechanisms remain unknown. In this study, we observed that the expression of SIRT6 increased during cementoblast differentiation initially. Analysis of the gain- and loss-of-function indicated that overexpressing SIRT6 in OCCM-30 cells suppresses cementoblast differentiation and mineralization and downregulating SIRT6 promotes cementogenesis. GLUT1, a glucose transporter necessary in cementogenesis, was inhibited by SIRT6. Overexpressing GLUT1 in SIRT6-overexpressed OCCM-30 cells rescued the inhibitory effect of SIRT6 on cementoblast differentiation and mineralization. Moreover, AMPK was activated after overexpressing SIRT6 and inhibited cementoblast differentiation and mineralization. Downregulating the expression of SIRT6 inhibited AMPK activity. Meanwhile, GLUT1 overexpression significantly decreased AMPK activity. Overall, on one hand, SIRT6 inhibited cementoblast differentiation and mineralization by suppressing GLUT1. On the other hand, SIRT6 inhibited cementoblast differentiation and mineralization by activating the AMPK pathway. GLUT1 overexpression also rescued the increased AMPK pathway activated by SIRT6.


Subject(s)
Cementogenesis , Dental Cementum/enzymology , Glucose Transporter Type 1/metabolism , Sirtuins/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Cell Differentiation , Cell Line , Dental Cementum/cytology , Glucose Transporter Type 1/genetics , Mice , Signal Transduction , Sirtuins/genetics , Up-Regulation
16.
J Cell Mol Med ; 22(8): 3899-3910, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29761910

ABSTRACT

Iroquois homeobox gene 5 (Irx5) is a highly conserved member of the Iroquois homeobox gene family. Members of this family play distinct and overlapping roles in normal embryonic cell patterning and development of malignancies. In this study, we observed that IRX5 was abnormally abundant in tongue squamous cell carcinoma (TSCC) tissues and cell lines. We used gain- and loss-of-function methods to overexpress and knockdown IRX5 expression in the TSCC cell line CAL27. Our results elucidated that elevated levels of IRX5 promoted proliferation, migration and invasion of TSCC cells, whereas stable or transient knockdown of IRX5 expression suppressed TSCC cell proliferation, migration and invasion. As a transcription factor, IRX5 performed this function by targeting osteopontin (OPN) promoter and activating the NF-κB pathway. Finally, studies in xenograft tumour model showed that IRX5 significantly enhanced OPN expression and promoted tumour growth. Taken together, our study elucidates a promotive effect of IRX5 in TSCC through the connection with OPN. These findings reveal the new molecular mechanism of TSCC, which may potentiate its use as a novel molecular therapy target for TSCC.

17.
J Dent ; 71: 43-48, 2018 04.
Article in English | MEDLINE | ID: mdl-29391182

ABSTRACT

OBJECTIVES: This study aims to apply electrophoretic deposition (EPD) for occlusion of dentinal tubules in vitro and investigate its effect on tubule occlusion and shear bond strength (SBS). METHODS: Charged mesoporous silica nanoparticles (MSNs) were synthesized and characterized through field-emission scanning electron microscopy (FESEM), transmission electron microscopy (TEM), dynamic light scattering (DLS), and Fourier transform infrared (FT-IR) spectroscopy analyses. Thirty-nine sensitive dentin specimens were modeled and assigned randomly to three groups with different treatments (n = 13 each): group 1, immersion in the MSN suspension; and groups 2 and 3, anodic EPD with the specimen on the negative and positive electrode respectively. The effect of dentinal tubule occlusion was evaluated by dentin permeability test (n = 10 each) and FESEM examination (n = 3 each). Moreover, 18 specimens were grouped (n = 6 each) and treated in the same method. A resin stick was bonded onto each of the specimen using a self-etch adhesive (single bond universal) for SBS testing. RESULTS: Negatively-charged MSNs were synthesized and characterized as small and well-dispersed particles. After the EPD treatment (group 3), the dentinal tubules were effectively occluded by MSNs, which infiltrated into the tubules at a depth of approximately 7-8 µm and tightly associated with the tubular inwalls. SBS was not significantly different among the three groups (P > 0.05). CONCLUSIONS: Synthesized MSNs were deposited into dentinal tubules by EPD treatment without compromising dentin bond strength. CLINICAL SIGNIFICANCE: Application of EPD is a new approach for occlusion of dentinal tubules and exhibits potential in the study of dentin hypersensitivity.


Subject(s)
Dental Occlusion , Dentin Sensitivity/drug therapy , Dentin/drug effects , Electrophoresis/methods , Bisphenol A-Glycidyl Methacrylate , Dentin/ultrastructure , Dentin Desensitizing Agents/chemistry , Dentin Permeability/drug effects , Dynamic Light Scattering , Electrodes , Humans , In Vitro Techniques , Materials Testing , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Nanoparticles/chemistry , Particle Size , Shear Strength , Silicon Dioxide/pharmacology , Surface Properties
18.
J Cell Physiol ; 233(3): 2213-2224, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28688217

ABSTRACT

Yes-associated protein 1 (YAP1) transcriptional coactivator is a mediator of mechanosensitive signaling. Cementum, which covers the tooth root surface, continuously senses external mechanical stimulation. Cementoblasts are responsible for the mineralization and maturation of the cementum. However, the effect of YAP1 on cementoblast differentiation remains largely unknown. In this study, we initially demonstrated that YAP1 overexpression enhanced the mineralization ability of cementoblasts. YAP1 upregulated the mRNA and protein expression of several cementogenesis markers, such as alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2), osteocalcin (OCN), and dentin matrix acidic phosphoprotein 1 (DMP1). The YAP1 overexpression group showed higher intensities of ALP and Alizarin red stain than the YAP1-knockdown group. Unexpectedly, a sharp increase in the expression of dentin sialophosphoprotein (DSPP) was induced by the overexpression of YAP1. Knockdown of YAP1 suppressed DSPP transcriptional activity. YAP1 overexpression activated Smad-dependent BMP signaling and slightly inhibited Erk1/2 signaling pathway activity. Treatment with specific BMP antagonist (LDN193189) prevented the upregulation of the mRNA levels of ALP, RUNX2, and OCN, as well as intensity of ALP-stained and mineralized nodules in cementoblasts. The Erk1/2 signaling pathway inhibitor (PD 98,059) upregulated these cementogenesis markers. Thus, our study suggested that YAP1 enhanced cementoblast mineralization in vitro. YAP1 exerted its effect on the cementoblast partly by regulating the Smad-dependent BMP and Erk1/2 signaling pathways.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone Morphogenetic Protein 1/metabolism , Cementogenesis/physiology , Dental Cementum/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Phosphoproteins/metabolism , Smad Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Alkaline Phosphatase/biosynthesis , Animals , Bone Morphogenetic Protein 1/antagonists & inhibitors , Cell Cycle Proteins , Cell Differentiation , Cell Line , Core Binding Factor Alpha 1 Subunit/biosynthesis , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Mice , Osteocalcin/biosynthesis , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sialoglycoproteins/biosynthesis , Sialoglycoproteins/genetics , YAP-Signaling Proteins
19.
Cell Physiol Biochem ; 44(6): 2174-2188, 2017.
Article in English | MEDLINE | ID: mdl-29241211

ABSTRACT

BACKGROUND/AIMS: Human dental pulp-derived mesenchymal stromal cells (hDPSCs) are promising seed cells for tissue engineering due to their easy accessibility and multi-lineage differentiation. Pannexin3 (Panx3) plays crucial roles during bone development and differentiation. The aim of the present study was to investigate the effect of Panx3 on osteogenesis of hDPSCs and the underlying mechanism. METHODS: Utilizing qRT-PCR, Western blot, and immunohistochemistry, we explored the change of Panx3 during osteogenic differentiation of hDPSCs. Next, hDPSCs with loss (Panx3 knockdown) and gain (Panx3 overexpression) of Panx3 function were developed to investigate the effects of Panx3 on osteogenic differentiation of hDPSC and the underlying mechanism. Finally, a commercial ß-TCP scaffold carrying Panx3-modified hDPSCs was utilized to evaluate bone defect repair. RESULTS: Panx3 was upregulated during osteogenic differentiation in a time-dependent manner. Panx3 overexpression promoted osteogenic differentiation of hDPSCs, whereas depletion of Panx3 resulted in a decline of differentiation, evidenced by upregulated expression of mineralization-related markers, increased alkaline phosphatase (ALP) activity, and enhanced ALP and Alizarin red staining. Panx3 was found to interact with the Wnt/ß-catenin signaling pathway, forming a negative feedback loop. However, Wnt/ß-catenin did not contribute to enhancement of osteogenic differentiation as observed in Panx3 overexpression. Moreover, Panx3 promoted osteogenic differentiation of hDPSCs via increasing ERK signaling pathway. Micro-CT and histological staining results showed that Panx3-modified hDPSCs significantly improved ossification of critical-sized bone defects. CONCLUSION: These findings suggest that Panx3 is a crucial modulator of hDPSCs differentiation.


Subject(s)
Connexins/genetics , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Osteogenesis , Skull/injuries , Up-Regulation , Adolescent , Adult , Animals , Cell Differentiation , Cells, Cultured , Connexins/metabolism , Dental Pulp/cytology , Fractures, Bone/pathology , Fractures, Bone/therapy , Gene Expression Regulation, Developmental , Humans , MAP Kinase Signaling System , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Rats , Rats, Sprague-Dawley , Skull/pathology , Wnt Signaling Pathway , Young Adult
20.
Int J Biochem Cell Biol ; 87: 104-113, 2017 06.
Article in English | MEDLINE | ID: mdl-28438716

ABSTRACT

YAP1 (Yes-associated protein 1) transcriptional coactivator is a downstream gene of the Hippo signaling pathway, which controls cell proliferation and differentiation. YAP1 plays a significant role in the regulation of cartilage and bone development. However, the molecular mechanism by which YAP1 regulates chondrocyte differentiation remains to be elucidated. Immunofluorescent staining was used to visualize the localization of YAP1 expression in the mouse chondroprogenitor ATDC5 cell line. ATDC5 cells with lentivirus-vector-mediated YAP1 overexpression and knockdown were established. The differentiation abilities were examined by real-time quantitative PCR and two staining methods The expression levels of sex-determining region Y-type high mobility group box protein (SOX9) and key proteins in the Wnt/ß-catenin pathway were analyzed by Western blot. The Dickkopf-1 (Dkk1) and small interfering RNA (siRNA) of ß-catenin were used for further study. The YAP1 protein was mainly expressed in the nucleus of ATDC5 cells. YAP1 overexpression enhanced chondrocyte proliferation but inhibited chondrocyte differentiation, which were contrary to the findings of the YAP1-knockdown group. Moreover, YAP1 overexpression activated Wnt/ß-catenin signaling pathway. Treatment with exogenous DKK1 and ß-catenin siRNA partially recaptured the effects of YAP1 overexpression on ATDC5 cell differentiation. Taken together, our study suggested that YAP1 attenuated ATDC5 cell chondrogenic and hypertrophic differentiation. We also demonstrated that YAP1 exerted its effect on the chondrocyte differentiation by activating the Wnt/ß-catenin signaling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation , Chondrocytes/cytology , Phosphoproteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cell Line , Cell Proliferation , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Hypertrophy/pathology , Mice , Phosphoproteins/deficiency , Phosphoproteins/genetics , Transcription Factors , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL