Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Small ; : e2311584, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38566551

ABSTRACT

2D materials have offered essential contributions to boosting biocatalytic efficiency in diverse biomedical applications due to the intrinsic enzyme-mimetic activity and massive specific surface area for loading metal catalytic centers. Since the difficulty of high-quality synthesis, the varied structure, and the tough choice of efficient surface loading sites with catalytic properties, the artificial building of 2D nanobiocatalysts still faces great challenges. Here, in this review, a timely and comprehensive summarization of the latest progress and future trends in the design and biotherapeutic applications of 2D nanobiocatalysts is provided, which is essential for their development. First, an overview of the synthesis-structure-fundamentals and structure-property relationships of 2D nanobiocatalysts, both metal-free and metal-based is provided. After that, the effective design of the active sites of nanobiocatalysts is discussed. Then, the progress of their applied research in recent years, including biomedical analysis, biomedical therapeutics, pharmacokinetics, and toxicology is systematically highlighted. Finally, future research directions of 2D nanobiocatalysts are prospected. Overall, this review to provide cutting-edge and multidisciplinary guidance for accelerating future developments and biomedical applications of 2D nanobiocatalysts is expected.

2.
Small ; : e2306966, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-38059865

ABSTRACT

Developing high-efficiency artificial biocatalysts for scavenging reactive oxygen species (ROS) is critical for treating inflammation diseases and promoting tissue regeneration. By mimicking the active sites in catalase, here, a Pt-clusters-equipped antioxidase-like biocatalysts (Pt─CN) with superior catalytic abilities for stem cell protection and periodontitis treatment are reported. Owing to the excellent effects of multiple Pt clusters, Pt─CN yields exceptional catalytic ROS-scavenging activities for multiple types of ROS. In vitro studies show that Pt─CN can effectively protect stem cell survival, adhesion, and differentiation in a high ROS levels microenvironment. Additionally, Pt─CN can reduce the M1/M2 ratio of macrophages when stimulated by lipopolysaccharide. In vivo treatment of mouse periodontitis further confirms the protection against bone loss and reduction in the inflammatory response. This study provides a basis for the application of biocatalysts with Pt catalytic center in macrophage polarization, stem cell protection, and periodontitis treatment, thus offering a new strategy for the design of high-performance artificial biocatalysts.

3.
Oral Dis ; 2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37771213

ABSTRACT

Semaphorin 3A (SEMA3A), also referred to as H-Sema III, is a molecule with significant biological importance in regulating physiological and pathological processes. However, its role in oral diseases, particularly its association with inflammatory immunity and alveolar bone remodeling defects, remains poorly understood. This comprehensive review article aims to elucidate the recent advances in understanding SEMA3A in the oral system, encompassing nerve formation, periodontitis, pulpitis, apical periodontitis, and oral squamous cell carcinoma. Notably, we explore its novel function in inflammatory immunomodulation and alveolar bone formation during oral infectious diseases. By doing so, this review enhances our comprehension of SEMA3A's role in oral biology and opens up possibilities for modulatory approaches and potential treatments in oral diseases.

4.
Small ; 19(50): e2303594, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37626465

ABSTRACT

Bacterial infection continues to be an increasing global health problem with the most widely accepted treatment paradigms restricted to antibiotics. However, the overuse and misuse of antibiotics have triggered multidrug resistance of bacteria, frustrating therapeutic outcomes, and leading to higher mortality rates. Even worse, the tendency of bacteria to form biofilms on living and nonliving surfaces further increases the difficulty in confronting bacteria because the extracellular matrix can act as a robust barrier to prevent the penetration of antibiotics and resist environmental damage. As a result, the inability to eliminate bacteria and biofilms often leads to persistent infection, implant failure, and device damage. Therefore, it is of paramount importance to develop alternative antimicrobial agents while avoiding the generation of bacterial resistance to prevent the large-scale growth of bacterial resistance. In recent years, nano-antibacterial materials have played a vital role in the antibacterial field because of their excellent physical and chemical properties. This review focuses on new physicochemical antibacterial strategies and versatile antibacterial nanomaterials, especially the mechanism and types of 2D antibacterial nanomaterials. In addition, this advanced review provides guidance on the development direction of antibiotic-free disinfections in the antibacterial field in the future.


Subject(s)
Anti-Infective Agents , Bacterial Infections , Nanostructures , Humans , Anti-Bacterial Agents/chemistry , Disinfection , Bacterial Infections/drug therapy , Bacteria
6.
Front Cell Dev Biol ; 11: 1209817, 2023.
Article in English | MEDLINE | ID: mdl-37457289

ABSTRACT

Mineralized tissues, such as teeth and bones, pose significant challenges for repair due to their hardness, low permeability, and limited blood flow compared to soft tissues. Bone morphogenetic proteins (BMPs) have been identified as playing a crucial role in mineralized tissue formation and repair. However, the application of large amounts of exogenous BMPs may cause side effects such as inflammation. Therefore, it is necessary to identify a more precise molecular target downstream of the ligands. Activin receptor-like kinase 3 (ALK3), a key transmembrane receptor, serves as a vital gateway for the transmission of BMP signals, triggering cellular responses. Recent research has yielded new insights into the regulatory roles of ALK3 in mineralized tissues. Experimental knockout or mutation of ALK3 has been shown to result in skeletal dysmorphisms and failure of tooth formation, eruption, and orthodontic tooth movement. This review summarizes the roles of ALK3 in mineralized tissue regulation and elucidates how ALK3-mediated signaling influences the physiology and pathology of teeth and bones. Additionally, this review provides a reference for recommended basic research and potential future treatment strategies for the repair and regeneration of mineralized tissues.

7.
Biomed Pharmacother ; 165: 115045, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37379643

ABSTRACT

Gene knockout is a technique routinely used in basic experimental research, particularly in mouse skeletal and developmental studies. Tamoxifen-induced Cre/loxp system is known for its temporal and spatial precision and commonly utilized by researchers. However, tamoxifen has been shown its side effects on affecting the phenotype of mouse bone directly. This review aimed to optimize tamoxifen administration regimens including its dosage and duration, to identify an optimal induction strategy that minimizes potential side effects while maintaining recombination efficacy. This study will help researchers in designing gene knockout experiments in bone when using tamoxifen.


Subject(s)
Integrases , Tamoxifen , Mice , Animals , Tamoxifen/pharmacology , Mice, Transgenic , Integrases/genetics , Gene Knockout Techniques
8.
ACS Nano ; 17(15): 15097-15112, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37378617

ABSTRACT

Periodontitis, one of the most common, challenging, and rapidly expanding oral diseases, is an oxidative stress-related disease caused by excessive reactive oxygen species (ROS) production. Developing ROS-scavenging materials to regulate the periodontium microenvironments is essential for treating periodontitis. Here, we report on creating cobalt oxide-supported Ir (CoO-Ir) as a cascade and ultrafast artificial antioxidase to alleviate local tissue inflammation and bone resorption in periodontitis. It is demonstrated that the Ir nanoclusters are uniformly supported on the CoO lattice, and there is stable chemical coupling and strong charge transfer from Co to Ir sites. Benefiting from its structural advantages, CoO-Ir presents cascade and ultrafast superoxide dismutase-catalase-like catalytic activities. Notably, it displays distinctly increased Vmax (76.249 mg L-1 min-1) and turnover number (2.736 s-1) when eliminating H2O2, which surpasses most of the by-far-reported artificial enzymes. Consequently, the CoO-Ir not only provides efficient cellular protection from ROS attack but also promotes osteogenetic differentiation in vitro. Furthermore, CoO-Ir can efficiently combat periodontitis by inhibiting inflammation-induced tissue destruction and promoting osteogenic regeneration. We believe that this report will shed meaningful light on creating cascade and ultrafast artificial antioxidases and offer an effective strategy to combat tissue inflammation and osteogenic resorption in oxidative stress-related diseases.


Subject(s)
Bone Resorption , Periodontitis , Humans , Reactive Oxygen Species , Hydrogen Peroxide , Periodontitis/drug therapy , Inflammation/drug therapy , Bone Resorption/drug therapy
9.
Angew Chem Int Ed Engl ; 62(22): e202302329, 2023 05 22.
Article in English | MEDLINE | ID: mdl-37002706

ABSTRACT

Constructing highly effective biocatalysts with controllable coordination geometry for eliminating reactive oxygen species (ROS) to address the current bottlenecks in stem-cell-based therapeutics remains challenging. Herein, inspired by the coordination structure of manganese-based antioxidase, we report a manganese-coordinated polyphthalocyanine-based biocatalyst (Mn-PcBC) with axial Mn-N5 sites and 2D d-π-conjugated networks that serves as an artificial antioxidase to rescue stem cell fate. Owing to the unique chemical and electronic structures, Mn-PcBC displays efficient, multifaceted, and robust ROS-scavenging activities, including elimination of H2 O2 and O2 ⋅- . Consequently, Mn-PcBC efficiently rescues the bioactivity and functionality of stem cells in high-ROS-level microenvironments by protecting the transcription of osteogenesis-related genes. This study offers essential insight into the crucial functions of axially coordinated Mn-N5 sites in ROS scavenging and suggests new strategies to create efficient artificial antioxidases for stem-cell therapies.


Subject(s)
Manganese , Stem Cells , Reactive Oxygen Species , Manganese/chemistry , Cell Differentiation
10.
Adv Mater ; 35(51): e2301836, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37089082

ABSTRACT

Metal alloys-structured electrocatalysts (MAECs) have made essential contributions to accelerating the practical applications of electrocatalytic devices in renewable energy systems. However, due to the complex atomic structures, varied electronic states, and abundant supports, precisely decoding the metal-metal interactions and structure-activity relationships of MAECs still confronts great challenges, which is critical to direct the future engineering and optimization of MAECs. Here, this timely review comprehensively summarizes the latest advances in creating the MAECs, including the metal-metal interactions, coordination microenvironments, and structure-activity relationships. First, the fundamental classification, design, characterization, and structural reconstruction of MAECs are outlined. Then, the electrocatalytic merits and modulation strategies of recent breakthroughs for noble and non-noble metal-structured MAECs are thoroughly discussed, such as solid solution alloys, intermetallic alloys, and single-atom alloys. Particularly, unique insights into the bond interactions, theoretical understanding, and operando techniques for mechanism disclosure are given. Thereafter, the current states of diverse MAECs with a unique focus on structural property-reactivity relationships, reaction pathways, and performance comparisons are discussed. Finally, the future challenges and perspectives for MAECs are systematically discussed. It is believed that this comprehensive review can offer a substantial impact on stimulating the widespread utilization of metal alloys-structured materials in electrocatalysis.

11.
Cell Commun Signal ; 21(1): 55, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36915160

ABSTRACT

Fibroblast growth factor 19 (FGF19) is recognized to play an essential role in cartilage development and physiology, and has emerged as a potential therapeutic target for skeletal metabolic diseases. However, FGF19-mediated cellular behavior in chondrocytes remains a big challenge. In the current study, we aimed to investigate the role of FGF19 on chondrocytes by characterizing mitochondrial biogenesis and fission-fusion dynamic equilibrium and exploring the underlying mechanism. We first found that FGF19 enhanced mitochondrial biogenesis in chondrocytes with the help of ß Klotho (KLB), a vital accessory protein for assisting the binding of FGF19 to its receptor, and the enhanced biogenesis accompanied with a fusion of mitochondria, reflecting in the elongation of individual mitochondria and the up-regulation of mitochondrial fusion proteins. We then revealed that FGF19-mediated mitochondrial biogenesis and fusion required the binding of FGF19 to the membrane receptor, FGFR4, and the activation of AMP-activated protein kinase alpha (AMPKα)/peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α)/sirtuin 1 (SIRT1) axis. Finally, we demonstrated that FGF19-mediated mitochondrial biogenesis and fusion was mainly dependent on the activation of p-p38 signaling. Inhibition of p38 signaling largely reduced the high expression of AMPKα/PGC-1α/SIRT1 axis, decreased the up-regulation of mitochondrial fusion proteins and impaired the enhancement of mitochondrial network morphology in chondrocytes induced by FGF19. Taking together, our results indicate that FGF19 could increase mitochondrial biogenesis and fusion via AMPKα-p38/MAPK signaling, which enlarge the understanding of FGF19 on chondrocyte metabolism. Video Abstract.


Subject(s)
AMP-Activated Protein Kinases , Transcription Factors , Transcription Factors/metabolism , AMP-Activated Protein Kinases/metabolism , Chondrocytes/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Organelle Biogenesis , Sirtuin 1/metabolism
12.
Regen Biomater ; 10: rbac100, 2023.
Article in English | MEDLINE | ID: mdl-36683745

ABSTRACT

Mechanical stiffness is recognized as a key physical factor and directs cell function via a mechanotransduction process, from extracellular physical cues to intracellular signaling cascades that affect transcriptional activity. Cells continually receive mechanical signals from both the surrounding matrix and adjacent cells. However, how mechanical stiffness cue at cell-substrate interfaces coordinates cell-cell junctions in guiding mesenchymal stem cell behaviors is poorly understood. Here, polydimethylsiloxane substrates with different stiffnesses were used to study mechanosensation/transduction mechanisms in controlling odontogenic differentiation of dental papilla cells (DPCs). DPC phenotypes (morphology and differentiation) changed in response to the applied force derived from stiff substrates. Significantly, higher expression of paxillin on stiffer substrates promoted DPC dentinogenesis. Upon treatment with siRNA to knockdown paxillin, N-cadherin increased mainly in the cytomembrane at the area of cell-cell contacts, whereas ß-catenin decreased in the nuclei. The result of a double luciferase reporter assay showed that stiffness promoted ß-catenin binding to TCF, which could coactivate the target genes associated with odontogenic differentiation, as evidenced by bioinformatics analysis. Finally, we determined that the addition of a ß-catenin inhibitor suppressed DPC mineralization in all the stiffness groups. Thus, our results indicated that a mechanotransduction process from cell-substrate interactions to cell-cell adhesions was required for DPC odontogenic differentiation under the stimulation of substrate stiffness. This finding suggests that stem cell fate specification under the stimulus of stiffness at the substrates is based on crosstalk between substrate interactions and adherens junctions, which provides an essential mechanism for cell-based tissue engineering.

13.
Cell Signal ; 105: 110605, 2023 05.
Article in English | MEDLINE | ID: mdl-36681291

ABSTRACT

Gap junction intercellular communication (GJIC) allows the transfer of material, message and energy between cells, which influences cell behaviors including cell proliferation, migration, differentiation and apoptosis and determines cell fate. Interleukin-10 (IL-10), a versatile cytokine, attracts more and more attention in the cartilage pathology such as osteoarthritis (OA) due to its potential in anti-inflammation and wound repair. However, whether IL-10 can mediate GJIC in chondrocytes remains elusive. In the current study, we aimed to explore the role of IL-10 on GJIC and its underlying mechanism. We found that IL-10 can promote GJIC in living chondrocytes. IL-10-enhanced GJIC in chondrocytes was dependent on the up-regulation of connexin 43 (Cx43). Knockdown experiment based on siRNA interference then confirmed that IL-10-enhanced GJIC required participation of IL-10 receptor 1 (IL-10R1). IL-10 activated signal transducer and activator of transcription 3 (STAT3) signaling and promoted the nuclear accumulation of p-STAT3 through IL-10 receptor 1. Inhibitor experiment further confirmed the importance of STAT3 signaling in IL-10-mediated GJIC. Taking together, our results provided a thorough process of IL-10-modulated cell-to-cell communication in chondrocytes and established a bridge between inflammatory factor, IL-10, and GJIC, which can increase our understanding about the physiology and pathology of cartilage.


Subject(s)
Chondrocytes , Interleukin-10 , Chondrocytes/metabolism , Interleukin-10/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Cell Communication , Receptors, Interleukin-10/metabolism
14.
Oral Dis ; 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36519511

ABSTRACT

OBJECTIVES: In vivo, the principal function of mechanosensitive odontoblasts is to synthesize and secrete the matrix which then calcifies and forms reactive dentin after exposure to appropriate stimuli. This study aims to develop the influence of mechanical factors on dentinogenesis based on odontoblasts, which contribute to reparative dentin formation. METHODS: We fabricated polydimethylsiloxane with different stiffnesses and seeded 17IIA11 odontoblast-like cells on the substrates in different stiffnesses. Cell morphology was detected by scanning electron microscope, and the mineralization phenotype was detected by alkaline phosphatase staining and alizarin red staining, while expression levels of dentinogenesis-related genes (including Runx2, Osx, and Alp) were assayed by qPCR. To explore mechanism, protein distribution and expression levels were detected by immunofluorescent staining, Western blotting, and immunoprecipitation. RESULTS: In our results, during dentinogenesis, 17IIA11 odontoblast-like cells appeared better extension on stiffer substrates. The binding between LAMB1 and FAK contributed to converting mechanical stimuli into biochemical signaling, thereby controlling mitogen-activated protein kinase kinase 1/2 activity in stiffness-driven dentinogenesis. CONCLUSION: The present study suggests odontoblast behaviors can be directly regulated by mechanical factors at cell-material interfaces, which offers fundamental mechanism in remodeling cell microenvironment, thereby contributing to physiological phenomena explanation and tissue engineering progress.

15.
Adv Mater ; 34(46): e2206208, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36065047

ABSTRACT

Exploring high-efficiency reactive oxygen species (ROS)-elimination materials is of great importance for combating oxidative stress in diverse diseases, especially stem-cell-based biotherapeutics. By mimicking the FeN active centers of natural catalase, here, an innovative concept to design ROS-elimination artificial biocatalysts with Ru catalytic centers for stem-cell protection is reported. The experimental studies and theoretical calculations have systematically disclosed the activity merits and structure diversities of different Ru sites when serving as ROS-elimination artificial biocatalysts. Benefiting from the metallic electronic structures and synergetic effects of multiple sites, the artificial biocatalysts with Ru cluster centers present exceptional ROS-elimination activity; notably, it shows much higher catalytic efficiency per Ru atom on decomposing H2 O2 when compared to the isolated single-atom Ru sites, which is more efficient than that of the natural antioxidants and recently reported state-of-the-art ROS-scavenging biocatalysts. The systematic stem-cell protection studies reveal that the catalase-like artificial biocatalysts can provide efficient rescue ability for survival, adhesion, and differentiation functions of human mesenchymal stem cells in high ROS level conditions. It is suggested that applying these artificial biocatalysts with Ru cluster centers will offer a new pathway for engineering high-performance ROS-scavenging materials in stem-cell-based therapeutics and many other ROS-related diseases.


Subject(s)
Cytoprotection , Oxidative Stress , Humans , Catalase/metabolism , Reactive Oxygen Species/metabolism , Catalysis
16.
Biochim Biophys Acta Mol Cell Res ; 1869(2): 119175, 2022 02.
Article in English | MEDLINE | ID: mdl-34863793

ABSTRACT

Connexin 43 (Cx43)-mediated gap junction intercellular communication (GJIC) plays a crucial role in the pathology and physiology of joint tissues. Transforming growth factor-ß2 (TGF-ß2), one of the potent regulatory factors in chondrocytes, plays a key role in the regulation of cell cycle and development of joint diseases. However, it is still unknown how TGF-ß2 mediates GJIC in chondrocytes. The aim of this study was to explore the potential mechanism by which TGF-ß2 regulates GJIC in chondrocytes. CCK-8 assays and scratch assays were performed to define the role of TGF-ß2 on cell proliferation and migration. The scrape loading/dye transfer assay and scanning electron microscopy (SEM) were used to verify the effect of TGF-ß2 on GJIC between chondrocytes. qPCR was performed to analyse the expression of genes in the gap junction protein family in chondrocytes. The expression of the Cx43 protein and phosphorylated Smad3 (p-Smad3) was evaluated by western blot assay. Immunofluorescence staining was used to explore p-Smad3 signalling pathway activation and Cx43 distribution. From these experiments, we found that the Cx43 protein was the most highly expressed member of the gap junction protein family in chondrocytes. We also found that TGF-ß2 facilitated cell-to-cell communication in chondrocytes by upregulating Cx43 expression in chondrocytes. Finally, we found that TGF-ß2 activated Smad3 signalling and promoted the nuclear aggregation of p-Smad3. Inhibition experiments by SIS3 also confirmed that TGF-ß2-mediated GJIC through p-Smad3 signalling. For the first time, this study confirmed that TGF-ß2 could regulate the formation of Cx43-mediated GJIC in chondrocytes via the canonical p-Smad3 signalling pathway.


Subject(s)
Cell Communication , Chondrocytes/metabolism , Connexin 43/metabolism , Gap Junctions/physiology , Smad3 Protein/metabolism , Transforming Growth Factor beta2/pharmacology , Animals , Cell Proliferation , Chondrocytes/cytology , Chondrocytes/drug effects , Connexin 43/genetics , Gap Junctions/drug effects , Mice , Mice, Inbred C57BL , Phosphorylation , Signal Transduction , Smad3 Protein/genetics
17.
Cell Biol Int ; 46(1): 34-45, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34643311

ABSTRACT

Wnts include more than 19 types of secreted glycoproteins that are involved in a wide range of pathological processes in oral and maxillofacial diseases. The transmission of Wnt signalling from the extracellular matrix into the nucleus includes canonical pathways and noncanonical pathways, which play an important role in tooth development, alveolar bone regeneration, and related diseases. In recent years, with the in-depth study of Wnt signalling in oral and maxillofacial-related diseases, many new conclusions and perspectives have been reached, and there are also some controversies. This article aims to summarise the roles of Wnt signalling in various oral diseases, including periodontitis, dental pulp disease, jaw disease, cleft palate, and abnormal tooth development, to provide researchers with a better and more comprehensive understanding of Wnts in oral and maxillofacial diseases.


Subject(s)
Mouth/metabolism , Periodontal Diseases/metabolism , Temporomandibular Joint Dysfunction Syndrome/metabolism , Tooth Diseases/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Dental Caries/genetics , Dental Caries/metabolism , Dental Caries/pathology , Gene Expression Regulation , Humans , Mouth/pathology , Odontogenesis , Periapical Periodontitis/genetics , Periapical Periodontitis/metabolism , Periapical Periodontitis/pathology , Periodontal Diseases/genetics , Periodontal Diseases/pathology , Pulpitis/genetics , Pulpitis/metabolism , Pulpitis/pathology , Temporomandibular Joint Dysfunction Syndrome/genetics , Temporomandibular Joint Dysfunction Syndrome/pathology , Tooth Diseases/genetics , Tooth Diseases/pathology , Wnt Proteins/genetics
18.
Heliyon ; 8(12): e12656, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36636224

ABSTRACT

Runt-related transcription factor-1 (Runx1) is well known for its functions in hematopoiesis and leukemia but recent research has focused on its role in skeletal development and osteoarthritis (OA). Deficiency of the Runx1 gene is fatal in early embryonic development, and specific knockout of Runx1 in cell lineages of cartilage and bone leads to delayed cartilage formation and impaired bone calcification. Runx1 can regulate genes including collagen type II (Col2a1) and X (Col10a1), SRY-box transcription factor 9 (Sox9), aggrecan (Acan) and matrix metalloproteinase 13 (MMP-13), and the up-regulation of Runx1 improves the homeostasis of the whole joint, even in the pathological state. Moreover, Runx1 is activated as a response to mechanical compression, but impaired in the joint with the pathological progress associated with osteoarthritis. Therefore, interpretation about the role of Runx1 could enlarge our understanding of key marker genes in the skeletal development and an increased understanding of Runx1 could be helpful to identify treatments for osteoarthritis. This review provides the most up-to-date advances in the roles and bio-mechanisms of Runx1 in healthy joints and osteoarthritis from all currently published articles and gives novel insights in therapeutic approaches to OA based on Runx1.

19.
Int J Biol Sci ; 17(14): 3862-3874, 2021.
Article in English | MEDLINE | ID: mdl-34671204

ABSTRACT

Fibroblast growth factors (FGFs) include a large family of growth factors that play a critical role in maintaining bone homeostasis, but the specific role of its members such as FGF7 does not well understand. Osteoblasts are a kind of major cells essential for bone formation. Osteoblasts interact with one another to create the unique structure of osteons. The well-connected osteons constitute the cortical bone. As an early osteocyte marker that triggers actin cytoskeleton dynamics, E11 is essential for osteoblasts' dendrites formation. However, the upstream which regulates E11 is mainly unknown. The purpose of this study was to examine the influence of FGF7 on the expression and the distribution of E11 in osteoblasts, which mediated osteoblasts' processes formation and gap junctional intercellular communication (GJIC) partly through connexin43 (Cx43). We first demonstrated that FGF7 increased the expression of E11 in osteoblasts. We then showed that FGF7 promoted osteoblasts' dendrites elongation and functional gap junctions formation. Furthermore, E11 interacted directly with Cx43 in primary osteoblasts. MAPK pathway and PI3K-AKT pathway were involved in the effect of FGF7. Our results shed light on the unique role of FGF7 on osteoblasts, which may indicate that FGF7 plays a more significant role in the later stages of bone development and homeostasis.


Subject(s)
Cell Communication/physiology , Connexin 43/metabolism , Fibroblast Growth Factor 7/physiology , Membrane Glycoproteins/metabolism , 3T3 Cells , Animals , Ligands , MAP Kinase Signaling System , Mice , Osteoblasts/metabolism
20.
Arch Oral Biol ; 131: 105264, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34598025

ABSTRACT

OBJECTIVE: Insulin-like growth factor 1 (IGF1) is one of the vital factors in regenerative endodontics. Previous studies have focused on the role of IGF1 in the mineralization of dental tissues. However, the role of IGF1 in the neural differentiation of dental stem cells was little discussed. DESIGN: IGF1 was overexpressed in human stem cells from the apical papilla (hSCAPs) by lentivirus and knocked down in hSCAPs by small interfering RNA. The neural differentiation level of hSCAPs was investigated histologically by HE staining and Nissl staining after neural induction for 3 days. The expression of proteins was examined by western blot and immunofluorescence. RESULTS: IGF1 promoted neural differentiation of hSCAPs, more cell processes and Nissl-positive body stained cells. IGF1 overexpression could both promote glial differentiation in hSCAPs, characterized by the increase of S100ß and GFAP proteins, and neuronal differentiation, characterized by the increase of ßIII-tubulin and functional GAD67/vGLUT1 proteins. Conversely, IGF1 knockdown suppressed both glial and neuronal differentiation. IGF1 activated AKT to regulate the early neural differentiation of hSCAPs. CONCLUSIONS: The results indicate IGF1 could promote neural differentiation of hSCAPs by activating AKT signaling and provide a cue for the candidate of induced neural seeding cells in regenerative endodontics.


Subject(s)
Cell Differentiation , Insulin-Like Growth Factor I , Stem Cells , Cells, Cultured , Dental Papilla/cytology , Humans , Lentivirus , Signal Transduction , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...