Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Int J Nanomedicine ; 19: 3143-3166, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38585472

RESUMO

Background: The ability of nanomaterials to induce osteogenic differentiation is limited, which seriously imped the repair of craniomaxillofacial bone defect. Magnetic graphene oxide (MGO) nanocomposites with the excellent physicochemical properties have great potential in bone tissue engineering. In this study, we aim to explore the craniomaxillofacial bone defect repairment effect of MGO nanocomposites and its underlying mechanism. Methods: The biocompatibility of MGO nanocomposites was verified by CCK8, live/dead staining and cytoskeleton staining. The function of MGO nanocomposites induced osteogenic differentiation of BMSCs was investigated by ALP activity detection, mineralized nodules staining, detection of osteogenic genes and proteins, and immune-histochemical staining. BMSCs with or without MGO osteogenic differentiation induction were collected and subjected to high-throughput circular ribonucleic acids (circRNAs) sequencing, and then crucial circRNA circAars was screened and identified. Bioinformatics analysis, Dual-luciferase reporter assay, RNA binding protein immunoprecipitation (RIP), fluorescence in situ hybridization (FISH) and osteogenic-related examinations were used to further explore the ability of circAars to participate in MGO nanocomposites regulation of osteogenic differentiation of BMSCs and its potential mechanism. Furthermore, critical-sized calvarial defects were constructed and were performed to verify the osteogenic differentiation induction effects and its potential mechanism induced by MGO nanocomposites. Results: We verify the good biocompatibility and osteogenic differentiation improvement effects of BMSCs mediated by MGO nanocomposites. Furthermore, a new circRNA-circAars, we find and identify, is obviously upregulated in BMSCs mediated by MGO nanocomposites. Silencing circAars could significantly decrease the osteogenic ability of MGO nanocomposites. The underlying mechanism involved circAars sponging miR-128-3p to regulate the expression of SMAD5, which played an important role in the repair craniomaxillofacial bone defects mediated by MGO nanocomposites. Conclusion: We found that MGO nanocomposites regulated osteogenic differentiation of BMSCs via the circAars/miR-128-3p/SMAD5 pathway, which provided a feasible and effective strategy for the treatment of craniomaxillofacial bone defects.


Assuntos
Grafite , MicroRNAs , Nanocompostos , MicroRNAs/genética , Osteogênese/genética , RNA Circular , Hibridização in Situ Fluorescente , Óxido de Magnésio , Células Cultivadas , Regeneração Óssea , Fenômenos Magnéticos , Diferenciação Celular
3.
Int J Nanomedicine ; 18: 797-812, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36814858

RESUMO

Purpose: Nanomaterial-based photodynamic therapy (PDT) has been commonly used for the treatment of cancerous tumors. Despite significant achievements made in this field, the intrinsic impact of nanomaterials-based PDT on the mechanical properties of oral squamous cell carcinoma (OSCC) cells is not entirely understood. Here, we used atomic force microscopy (AFM) to measure the stiffness of OSCC cells subjected to PDT in co-culture systems to evaluate the T cell-mediated cancer cell-killing effects. Methods: In this study, AFM was used to assess the stiffness of PDT-subjected cells. The phototoxicity of graphdiyne oxide (GDYO) was assessed using confocal laser scanning microscopy (CLSM), measurements of membrane cholesterol levels, and assessments of the F-actin cytoskeleton. A co-culture system was used to evaluate the effects of CD8+ T cells (cytotoxic T lymphocytes), demonstrating how PDT modulates the mechanical properties of cancer cells and activates T cell responses. The antitumor immunotherapeutic effect of GDYO was further evaluated in a murine xenograft model. Results: GDYO increased the mechanical stiffness of tumor cells and augmented T-cell cytotoxicity and inflammatory cytokine secretion (IFN-γ and TNF-α) under laser in vitro. Furthermore, GDYO-based PDT exerted inhibitory effects on OSCC models and elicited antitumor immune responses via specific cytotoxic T cells. Conclusion: These results highlight that GDYO is a promising candidate for OSCC therapy, shifting the mechanical forces of OSCC cells and breaking through the barriers of the immunosuppressive tumor microenvironment. Our study provides a novel perspective on nanomaterial-based antitumor therapies.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Fotoquimioterapia , Humanos , Animais , Camundongos , Carcinoma de Células Escamosas/patologia , Linfócitos T CD8-Positivos , Óxidos , Fotoquimioterapia/métodos , Neoplasias Bucais/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço , Imunidade , Linhagem Celular Tumoral , Microambiente Tumoral
4.
Acta Biomater ; 159: 338-352, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36669551

RESUMO

There is growing interest in the effect of innate immune silencing in "cold" tumors, which always fail in the immune checkpoint blockade monotherapy using PD-L1 monoclonal antibodies (aPD-L1). Combination of aPD-L1 with photodynamic therapy, i.e., photoimmunotherapy, is a promising strategy to improve the mono immunotherapy. Nuclear-targeting nanoparticles could elicit a type I interferon (IFN)-mediated innate immune response and reverse the immunosuppressive microenvironment for long-term immunotherapy of "cold" tumors. Photosensitizers such as zinc phthalocyanine (ZnPc) have limited ability to target the nucleus and activate innate sensing pathways to minimize tumor recurrence. Additionally, the relationship between nanoparticle size and nuclear entry capacity remains unclear. Herein, graphene quantum dots (GQDs) were employed as aPD-L1 and ZnPc carriers. Three particle sizes (200 nm, 32 nm and 5 nm) of aPD-L1/ZnPc/GQD-PEG (PZGE) were synthesized and tested. The 5 nm nanoparticles achieved the best nuclear enrichment capacity contributing to their ultrasmall size. Notably, 5 nm PZGE-based photodynamic therapy enabled an amplification of the type I IFN-mediated innate immune response and could convert "immune-cold" tumors into "immune-hot" ones. Utilizing their size advantage to target the nucleus, 5 nm nanoparticles induced DNA damage and activated the type I IFN-mediated innate immune response, subsequently promoting cytotoxic T-lymphocyte infiltration and reversing negative PD-L1 expression. Furthermore, the nanoplatform we designed is promising for the effective suppression of distant oral squamous cell carcinoma. Thus, for the first time, this study presents a size design strategy for nuclear-targeted photo-controlled immune adjuvants and the nuclear-targeted phototherapy-mediated immunomodulatory functions of type I IFN innate immune signalling for "immune-cold" tumors. STATEMENT OF SIGNIFICANCE: The potential of commonly used photosensitizers to activate innate sensing pathways for producing type I IFNs is limited due to the lack of nuclear targeting. Facilitating the nuclear-targeting of photosensitizers to enhance innate immune response and execute long-term tumor killing effect would be a promising strategy for "cold" tumor photoimmunotherapy. Herein, we report an optimal size of PZGE nanoparticles that enable the nuclear-targeting of ZnPc, which reinforces the type I IFN-mediated innate immune response, synergistically reversing "cold tumors" to "hot tumors" for effective primary and distant tumor photoimmunotherapy. This work highlights the marked efficacy of ultrasmall nuclear-located nanocarriers and offers new insight into "immune-cold tumors" via prominent innate immune activation mediated by nuclear-targeting photoimmunotherapy.


Assuntos
Carcinoma de Células Escamosas , Interferon Tipo I , Neoplasias Bucais , Neoplasias , Humanos , Antígeno B7-H1 , Linhagem Celular Tumoral , Recidiva Local de Neoplasia , Neoplasias/terapia , Fármacos Fotossensibilizantes , Fototerapia , Microambiente Tumoral , Imunoterapia
5.
Mater Sci Eng C Mater Biol Appl ; 131: 112499, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34857285

RESUMO

Porous Ti6Al4V scaffolds are characterized by high porosity, low elastic modulus, and good osteogenesis and vascularization, which are expected to facilitate the repair of large-scale bone defects in future clinical applications. Ti6Al4V scaffolds are divided into regular and irregular structures according to the pore structure, but the pore structure more capable of promoting bone regeneration and angiogenesis has not yet been reported. The purpose of this study was to explore the optimal pore structure and pore size of the Ti6Al4V porous scaffold for the repair of large-area bone defects and the promotion of vascularization in the early stage of osteogenesis. 7 groups of porous Ti6Al4V scaffolds, named NP, R8, R9, R10, P8, P9 and P10, were fabricated by Electron-beam-melting (EBM). Live/dead staining, immunofluorescence staining, SEM, CCK8, ALP, and PCR were used to detect the adhesion, proliferation, and differentiation of BMSCs on different groups of scaffolds. Hematoxylin-eosin (HE) staining and Van Gieson (VG) staining were used to detect bone regeneration and angiogenesis in vivo. The research results showed that as the pore size of the scaffold increased, the surface area and volume of the scaffold gradually decreased, and cell proliferation ability and cell viability gradually increased. The ability of cells to vascularize on scaffolds with irregular pore sizes was stronger than that on scaffolds with regular pore sizes. Micro-CT 3D reconstruction images showed that bone regeneration was obvious and new blood vessels were thick on the P10 scaffold. HE and VG staining showed that the proportion of bone area on the scaffolds with irregular pores was higher than that on scaffolds with regular pores. P10 had better mechanical properties and were more conducive to bone tissue ingrowth and blood vessel formation, thereby facilitating the repair of large-area bone defects.


Assuntos
Regeneração Óssea , Alicerces Teciduais , Ligas , Osteogênese , Porosidade , Titânio
7.
Exp Cell Res ; 408(2): 112864, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34626586

RESUMO

Dental implant surgery is currently a routine therapy for the repair of missing dentition or dentition defects. Both clinical and basic research have elucidated that oxidative stress caused by the accumulation of reactive oxygen species (ROS) for various reasons impairs the process of osteointegration after dental implantation. Therefore, the osteogenic micro-environment must be ameliorated to decrease the damage caused by oxidative stress. Selenomethionine (SEMET) has been reported to play an important role in alleviating oxidative stress and accelerating cell viability and growth. However, it remains unclear whether it exerts protective effects on bone-marrow-derived mesenchymal stem cells (BMSCs) under oxidative stress. In this study, we explored the influence of selenomethionine on the viability and osteogenic differentiation of BMSCs under oxidative stress and the underlying mechanisms. Results showed that 1 µM selenomethionine was the optimum concentration for BMSCs under H2O2 stimulation. H2O2-induced oxidative stress suppressed the viability and osteogenic differentiation of BMSCs, manifested by the increases in ROS production and cell apoptosis rates, and by the decrease of osteogenic differentiation-related markers. Notably, the aforementioned oxidative damage and osteogenic dysfunction induced by H2O2 were rescued by selenomethionine. Furthermore, we found that the PTEN expression level was suppressed and its downstream PI3K/AKT pathway was activated by selenomethionine. However, when PTEN was stimulated, the PI3K/AKT pathway was down-regulated, and the protective effects of selenomethionine on BMSC osteogenic differentiation diminished, while the inhibition of PTEN up-regulated the protective effects of selenomethionine. Together, these results revealed that selenomethionine could attenuate H2O2-induced BMSC dysfunction through an antioxidant effect, modulated via the PTEN/PI3K/AKT pathway, suggesting that selenomethionine is a promising antioxidant candidate for reducing oxidative stress during the process of dental implant osteointegration.


Assuntos
Antioxidantes/farmacologia , Diferenciação Celular/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Selenometionina/farmacologia , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Implantes Dentários/efeitos adversos , Humanos , Peróxido de Hidrogênio/toxicidade , Células-Tronco Mesenquimais/efeitos dos fármacos , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
8.
J Cancer ; 12(20): 5999-6011, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34539874

RESUMO

Background: Malignant proliferation and cervical lymphatic metastasis restrict the prognosis of oral squamous cell carcinoma (OSCC). Erythropoietin-producing human hepatocellular B4 (EPHB4) regulates a series of tumour functions involving tumourigenesis, cancer cell attachment and metastasis. However, the mechanism of EphB4 regulating the malignant progression of OSCC has not been fully elucidated. Methods: EPHB4 expression was analysed in 65 OSCC samples and adjacent noncancerous tissues through immunohistochemistry (IHC). siRNA and overexpression plasmids were transfected into OSCC cells to modify EPHB4 expression, and then, regulatory functions were explored in vitro and in vivo. Co-immunoprecipitation (Co-IP) and mass spectrometry were applied to detect proteins interacting with EPHB4. Subsequently, protein stability assays and NF-κB pathway inhibition assays were used to verify the regulation of EPHB4, high-mobility group box 1 (HMGB1) and nuclear factor-κB (NF-κB) activation. Results: EPHB4 was found to be highly expressed in OSCC tissues, which was related to tumour stage and lymphatic metastasis and resulted in a poor prognosis. Cellular experiments and mouse tongue xenograft models further confirmed that high EPHB4 expression promoted the proliferation and metastasis of OSCC tumours. Mechanistically, co-IP and mass spectrometry studies indicated that EPHB4 could bind to HMGB1 and maintain HMGB1 stability. Downregulation of HMGB1 inhibited the proliferation and metastasis of OSCC cells and inhibited NF-κB phosphorylation activation but did not affect EPHB4 expression. Conclusion: This study revealed the mechanism by which EPHB4 promotes the proliferation and metastasis of OSCC by activating the HMGB1-mediated NF-κB signalling pathway, which can be exploited as a novel marker or therapeutic target to control metastasis and improve the survival rate of OSCC.

9.
ACS Biomater Sci Eng ; 7(10): 4933-4945, 2021 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-34583510

RESUMO

Titanium and its alloys have been widely used as bone implants, but for reduced treatment span, improvements are urgently needed to achieve faster and better osteointegration. In this study, we found that miR-132-3p inhibited bone-marrow-derived stem cell (BMSC) osteogenic differentiation via targeting BMP2, and that inhibiting miR-132-3p could significantly improve the osteogenic capability of BMSCs. Moreover, we fabricated a biocompatible selenomethionine (SEMET)-modified polyethylene glycol (PEG)/polyethylenimine (PEI) nanoparticle (SeNP) cross-linked with 0.2% gelatin solutions and delivered miR-132-3p inhibitor to biofunctionalize alkali heat-treated titanium implants, resulting in the development of a novel coating for reverse transfection. The biological performances of PEG/PEI/miR-132-3p inhibitor and SeNP/miR-132-3p inhibitor-biofunctionalized titanium were compared. The biological effects, including cell viability, cytotoxicity, adhesion, cellular uptake, and osteogenic capacity of SeNP/miR-132-3p inhibitor-biofunctionalized titanium implants, were then assessed. Results showed that SeNPs presented appropriate morphology, diameter, and positive zeta potential for efficient gene delivery. The transfection efficiency of the SeNP/miR-132-3p inhibitor was comparable to that of the PEG/PEI/miR-132-3p inhibitor, but the former induced less reactive oxygen species (ROS) production and lower apoptosis rates. Confocal laser scanning microscopy (CLSM) demonstrated that SeNP/miR-132-3p inhibitor nanoparticles released from the titanium surfaces and were taken up by adherent BMSCs. In addition, the release profile showed that transfection could obtain a long-lasting silencing effect for more than 2 weeks. The cell viability, cytotoxicity, and cell spreading of SeNP/miRNA-132-3p inhibitor-biofunctionalized titanium were comparable with those of untreated titanium and the SeNP/miRNA-132-3p inhibitor negative control (NC)-biofunctionalized titanium but resulted in higher ALP activity and osteogenic gene expression levels. In vivo animal studies further certified that SeNP/miRNA-132-3p inhibitor nanoparticles from titanium surfaces promoted osteointegration, which was revealed by microcomputed tomography (micro-CT) and histological observations. Taken together, these findings suggested that selenomethionine-modified PEI-based nanoparticles could achieve better biocompatibility. Moreover, titanium implants biofunctionalized by SeNP/miRNA-132-3p inhibitor nanoparticles might have significant clinical potential for more effective osteointegration.


Assuntos
MicroRNAs , Nanopartículas , Animais , MicroRNAs/genética , Osteogênese , Polietilenoimina , Selenometionina , Titânio , Microtomografia por Raio-X
10.
Angew Chem Int Ed Engl ; 60(44): 23608-23613, 2021 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-34459532

RESUMO

Herein, we report the first example of using mesoporous hydrogen-bonded organic frameworks (MHOFs) as the protecting scaffold to organize a biocatalytic cascade. The confined microenvironment of MHOFs has robust and large transport channels, allowing the efficient transport of a wide range of biocatalytic substrates. This new MHOF-confined cascade system shows superior activity, extended scope of catalytic substrates, and ultrahigh stability that enables the operation of complex chemical transformations in a porous carrier. In addition, the advantages of MHOF-confined cascades system for point-of-care biosensing are also demonstrated. This study highlights the advantages of HOFs as scaffold for multiple enzyme assemblies, which has huge potential for mimicking complex cellular transformation networks in a controllable manner.


Assuntos
Técnicas Biossensoriais , Glucose Oxidase/metabolismo , Peroxidase do Rábano Silvestre/metabolismo , Imidazóis/metabolismo , Sistemas Automatizados de Assistência Junto ao Leito , Zeolitas/metabolismo , Biocatálise , Enzimas Imobilizadas/química , Enzimas Imobilizadas/metabolismo , Glucose Oxidase/química , Peroxidase do Rábano Silvestre/química , Ligação de Hidrogênio , Imidazóis/química , Tamanho da Partícula , Porosidade , Propriedades de Superfície , Zeolitas/química
11.
BMC Cancer ; 21(1): 835, 2021 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-34284753

RESUMO

BACKGROUND: The prognosis of oral squamous cell carcinoma (OSCC) patients is difficult to predict or describe due to its high-level heterogeneity and complex aetiologic factors. Ferroptosis is a novel form of iron-dependent cell death that is closely related to tumour growth and progression. This study aims to clarify the predictive value of ferroptosis-related genes (FRGs) on the overall survival(OS) of OSCC patients. METHODS: The mRNA expression profile of FRGs and clinical information of patients with OSCC were collected from the TCGA database. Candidate differentially expressed ferroptosis-related genes (DE-FRGs) were identified by analysing differences between OSCC and adjacent normal tissues. A gene signature of prognosis-related DE-FRGs was established by univariate Cox analysis and LASSO analysis in the training set. Patients were then divided into high- and low-risk groups according to the cut-off value of risk scores, A nomogram was constructed to quantify the contributions of gene signature and clinical parameters to OS. Then several bioinformatics analyses were used to verify the reliability and accuracy of the model in the validation set. Finally, single-sample gene set enrichment analysis (ssGSEA) was also performed to reveal the underlying differences in immune status between different risk groups. RESULTS: A prognostic model was constructed based on 10 ferroptosis-related genes. Patients in high-risk group had a significantly worse OS (p < 0.001). The gene signature was verified as an independent predictor for the OS of OSCC patients (HR > 1, p < 0.001). The receiver operating characteristic curve displayed the favour predictive performance of the risk model. The prediction nomogram successfully quantified each indicator's contribution to survival and the concordance index and calibration plots showed its superior predictive capacity. Finally, ssGSEA preliminarily indicated that the poor prognosis in the high-risk group might result from the dysregulation of immune status. CONCLUSION: This study established a 10-ferroptosis-releated gene signature and nomogram that can be used to predict the prognosis of OSCC patients, which provides new insight for future anticancer therapies based on potential FRG targets.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/genética , Ferroptose/genética , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/genética , Neoplasias Bucais/genética , Carcinoma de Células Escamosas/patologia , Humanos , Pessoa de Meia-Idade , Neoplasias Bucais/patologia , Prognóstico
12.
Nanomedicine ; 37: 102435, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34186257

RESUMO

The biological behaviors of magnetic graphene oxide (MGO) in a static magnetic field (SMF) are unknown. The current study is to investigate the cellular behaviors, osteogenesis and the mechanism in BMSCs treated with MGO combined with an SMF. Results showed that the synthetic MGO particles were bio-compatible and could significantly improve the osteogenesis of BMSCs under SMFs, as verified by elevated alkaline phosphatase activity, mineralized nodule formation, and expressions of mRNA and protein levels. Under SMF at the same intensity, the addition of graphene oxide to Fe3O4 could increase the osteogenic ability of BMSCs. The Wnt/ß-catenin pathway was indicated to be related to the MGO-driven osteogenic behavior of the BMSCs under SMF. Taken together, our findings suggested that MGO under an SMF could promote osteogenesis in BMSCs through the Wnt/ß-catenin pathway and hence should attract more attention for practical applications in bone tissue regeneration.


Assuntos
Grafite/farmacologia , Campos Magnéticos , Nanopartículas de Magnetita/química , Osteogênese/genética , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos da radiação , Grafite/química , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos da radiação , Osteogênese/efeitos dos fármacos , Osteogênese/efeitos da radiação , Ratos , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/efeitos da radiação
13.
Oncogene ; 40(20): 3548-3563, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33927350

RESUMO

Mitochondrial oxidative phosphorylation (OXPHOS) is a vital regulator of tumor metastasis. However, the mechanisms governing OXPHOS to facilitate tumor metastasis remain unclear. In this study, we discovered that arginine 21(R21) and lysine 108 (K108) of mitochondrial ribosomal protein S23 (MRPS23) was methylated by the protein arginine methyltransferase 7 (PRMT7) and SET-domain-containing protein 6 (SETD6), respectively. R21 methylation accelerated the poly-ubiquitin-dependent degradation of MRPS23 to a low level. The MRPS23 degradation inhibited OXPHOS with elevated mtROS level, which consequently increased breast cancer cell invasion and metastasis. In contrast, K108 methylation increased MRPS23 stability, and K108 methylation coordinated with R21 methylation to maintain a low level of MRPS23, which was in favor of supporting breast cancer cell survival through regulating OXPHOS. Consistently, R21 and K108 methylation was correlated with malignant breast carcinoma. Significantly, our findings unveil a unique mechanism of controlling OXPHOS by arginine and lysine methylation and point to the impact of the PRMT7-SETD6-MRPS23 axis during breast cancer metastasis.


Assuntos
Arginina/química , Neoplasias da Mama/metabolismo , Lisina/química , Mitocôndrias/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Feminino , Humanos , Metilação , Mitocôndrias/patologia , Metástase Neoplásica , Organoides , Fosforilação Oxidativa , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Processamento de Proteína Pós-Traducional , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo
14.
Int J Nanomedicine ; 15: 9627-9638, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33293811

RESUMO

INTRODUCTION: With the innovation of photosensitizers, photodynamic therapy is now widely used in antitumor detection and treatment. Graphene quantum dots (GQDs) are proposed as a promising alternative photosensitizer due to their high biocompatibility, specific photoactivity, and strong tumor concentration. However, the changes in host immunity triggered by GQDs have only rarely been reported. METHODS: In this work, GQDs as photosensitizers were conjugated to polyethylene glycol (PEG) to enhance solubility and blood circulation. The phototoxicity of the resulting GQD-PEG nanomaterials was then detected in vitro and in vivo. The antitumor immunity triggered by GQD-PEG under irradiation was further evaluated in an oral squamous cell carcinoma animal model. RESULTS: The obtained GQD-PEG nanomaterials exhibited low cytotoxicity, good solution stability, and excellent endocytosis. Both in vitro and in vivo, all demonstrated strong ablation for oral squamous cell carcinoma under irradiation. Meanwhile, host-immunity-related CD8+ T cells (cytotoxic T lymphocytes) and proinflammatory cytokines, including IFN-γ and TNF-α, were significantly increased after photo-activated antitumor activity. CONCLUSION: These results highlight the dominant role of GQD-PEG in photodynamic therapy and could have significant implications for further combination therapy as a promising antitumor immune response strategy triggered by nanomaterials.


Assuntos
Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/imunologia , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/imunologia , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linhagem Celular Tumoral , Grafite/química , Humanos , Camundongos Endogâmicos C3H , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacocinética , Pontos Quânticos/química , Distribuição Tecidual
15.
ACS Omega ; 5(44): 28684-28692, 2020 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-33195921

RESUMO

Ti6Al4V is widely used in implants in the fields of orthopedics and dentistry due to its high compressive strength and good biocompatibility. Nevertheless, Ti6Al4V has a certain degree of biological inertness and the elastic modulus of Ti6Al4V is much higher than the cortex and trabecular bone. In this study, we designed and printed a new type of pore size Ti6Al4V with like-trabecular structure scaffold (the pore size is 800/900/1000 µm, named P8/P9/P10, respectively) with electron beam melting (EBM). Its elastic modulus, compressive strength, and other physical and chemical properties, as well as cell adhesion, proliferation, and differentiation ability and in vitro biological properties were studied. The physical and chemical performance test results showed that as the pore size increased, the surface wettability increased and the elastic modulus decreased. As the pore size increased, F-actin and alkaline phosphatase (ALP) increased significantly, and osteogenesis-related genes including BMP2, OCN, RUNX2, and ALP were upregulated significantly. The reason may be that the components on the Ti6Al4V pore size may have an influence on intracellular signal conversion and then change the mode of cell proliferation and diffusion. In summary, the like-trabecular porous structure can effectively reduce the elastic modulus of metal materials, thereby avoiding stress concentration and promoting the adhesion and proliferation of osteoblasts. Porous materials with larger pores are more conducive to the proliferation and differentiation of osteoblasts. The irregular porous Ti6Al4V scaffold prepared by the EBM technology has good mechanical properties and the potential to promote adhesion, proliferation, and differentiation of osteoblasts, and has the possibility of application in the field of implantation.

16.
Mol Ther Nucleic Acids ; 19: 961-973, 2020 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-32032888

RESUMO

Accumulating evidence has demonstrated that circular RNAs (circRNAs) play important roles in regulating gene expression involved in tumor development. However, the role of circRNAs in modulating the radiosensitivity of oral squamous cell carcinoma (OSCC) and its potential mechanisms have not been documented. We performed high-throughput RNA sequencing (RNA-seq) to investigate the circRNA expression profile in OSCC patients and discovered that the circATRNL1 expression was significantly downregulated and closely related to tumor progression. The circATRNL1 was structurally validated via Sanger sequencing, RNase R treatment, and specific convergent and divergent primer amplification. Importantly, the expression levels of circATRNL1 decreased after irradiation treatment, and upregulation of circATRNL1 enhanced the radiosensitivity of OSCC through suppressing proliferation and the colony survival fraction, inducing apoptosis and cell-cycle arrest. Moreover, we observed that circATRNL1 could directly bind to microRNA-23a-3p (miR-23a-3p) and relieve inhibition for the target gene PTEN. In addition, the tumor radiosensitivity-promoting effect of circATRNL1 overexpression was blocked by miR-23a-3p in OSCC. Further experiments also showed that PTEN can reverse the inhibitory effect of OSCC radiosensitivity triggered by miR-23a-3p. We concluded that circANTRL1 may function as the sponge of miR-23a-3p to promote PTEN expression and eventually contributes to OSCC radiosensitivity enhancement. This study indicates that circANTRL1 may be a novel therapeutic target to improve the efficiency of radiotherapy in OSCC.

17.
J Biomed Mater Res A ; 108(3): 614-624, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31742875

RESUMO

This study was aimed to investigate the toxic effects of pristine graphene oxide (GO) nanosheets on bone-marrow-derived mesenchymal stem cells (BMSCs), a type of traditional seed cells in tissue regeneration engineering. First, a GO suspension was prepared and characterized by transmission electron microscopy, Fourier transform infrared spectroscopy, and Raman shifts. Then, rat BMSCs were isolated and characterized. Subsequently, cell proliferation, membrane integrity, cell cycle, cell apoptosis, mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) were measured. In addition, relevant proteins of the mitochondrial apoptotic pathway and autophagy were analyzed. Our results showed that a high concentration of GO inhibited cell viability and membrane integrity, while cell apoptosis and cell-cycle arrest were induced by GO. Further, GO significantly increased ROS generation and MMP loss with an upregulation of Cleaved Caspase-3, LC3-II/I, and Beclin-1 and a downregulation of Bcl-2 and Caspase3. We concluded that the toxic effects of GO on BMSCs occurred in a dose-dependent manner via the mitochondrial apoptotic pathway and autophagy.


Assuntos
Materiais Biocompatíveis/toxicidade , Regeneração Óssea , Grafite/toxicidade , Células-Tronco Mesenquimais/citologia , Nanoestruturas/toxicidade , Animais , Apoptose/efeitos dos fármacos , Materiais Biocompatíveis/química , Regeneração Óssea/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Grafite/química , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Nanoestruturas/química , Ratos Sprague-Dawley
18.
J Biomed Mater Res A ; 108(1): 50-60, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31443121

RESUMO

The scaffold-free cell sheet plays an important role in stem-cell-based regeneration. Graphene oxide (GO) endows nanoparticles (NPs) with special characteristics and therefore has attracted increasing attention in recent years. However, the existence of toxicity in GO and its derivatives limits their ability to promote osteogenic differentiation. Magnetic graphene oxide (MGO), a novel combination of Fe3 O4 and GO with diverse unique properties, has not been studied in bone tissue engineering. In this study, MGO was fabricated, and the previously undiscovered relationships-including cellular behavior and the effects of osteogenic differentiation and related mechanisms of MGO in rat bone-marrow-derived mesenchymal stem cells (BMSCs)-were investigated for the first time. Here, we found that MGO was not only biocompatible at low concentrations, but also could significantly accelerate osteogenic differentiation in BMSCs. Both the cellular behavior and bone-formation differentiation in BMSCs treated with MGO showed concentration-dependent characteristics. In addition, the regulation of osteogenic differentiation in BMSCs treated with MGO might be involved with the Wnt/ß-catenin and BMP signaling pathways. Furthermore, MGO demonstrated a better ability for osteogenic differentiation in BMSCs than did GO. The current work indicated a significant use for MGO nanocomposite scaffolds in biocompatibility and bone regeneration, which could provide new insight into bone regeneration in the future.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Grafite/farmacologia , Fenômenos Magnéticos , Células-Tronco Mesenquimais/citologia , Osteogênese/efeitos dos fármacos , Fosfatase Alcalina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Contagem de Células , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Nanopartículas/química , Nanopartículas/ultraestrutura , Necrose , Osteogênese/genética , Ratos Sprague-Dawley
19.
Transl Cancer Res ; 9(4): 2683-2691, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35117627

RESUMO

BACKGROUND: Lung cancer is the most common and lethal tumor in the world, and the number of patients who die from lung cancer is growing steadily. Because of conventional chemotherapy drugs' poor tumor selectivity, side effects are significant. Conducting relevant studies and developing highly efficient and low toxicity anti-cancer drugs are urgently needed. Dendrobium officinale, which belongs to Orchidaceae aerophyte, has the characteristic of slow growth and lower natural propagation rate. In China, Dendrobium officinale has a very high value and is often referred to as the "gold of herbs". According to reports in the literature, the active ingredients of Dendrobium officinale have anticancer activity and inhibit neovascularization's potential. This study aimed to investigate the inhibitory effect of Dendrobium officinale in A549 lung cancer cells and its potential involvement in slowing tumor growth. METHODS: We cultured A549 cells and established a cancer xenograft model in nude mice. Infused stomach with Dendrobium officinale was applied to the nude mouse model. Tumor volume and body weight were recorded. RESULTS: The results show that, compared with the negative control group, the gross tumor volume (GTV) of treatment groups decreased (all P<0.05), while the effect of the high concentration of the Dendrobium officinale was more significant than that found in the medium and low group. We believe that Dendrobium officinale exhibits a promising antitumor effect in the nude mouse tumor model. The best treatment concentrations for the nude mouse tumor model were achieved when treatment with the drug began about 7-15 days, and was more significant in high concentrations. CONCLUSIONS: Dendrobium officinale has potent effects of inhibiting tumor on the nude mouse tumor model.

20.
J Trace Elem Med Biol ; 55: 64-70, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31345368

RESUMO

BACKGROUND: Patients with metabolic bone diseases often have high risk of titanium implant failure due to compromised bone regeneration ability. Clinical evidence indicates that the poor osteogenic ability is partly because of excessive oxidative stress. To date, specific treatments for these patients are urgently needed. Ebselen, a non-toxic organoselenium compound, is reported to be a potent antioxidant agent. In this study, we hypothesized that ebselen exerted protective effects on osteogenic differentiation of bone-marrow-derived mesenchymal stem cells (BMSCs) under oxidative stress. METHODS: BMSCs were isolated from SD rats, and their morphology and multiple differentiation abilities were characterized. Proliferation rates of BMSCs treated with different concentrations of ebselen were analyzed. Then BMSCs were pretreated by hydrogen peroxide (H2O2), after which ebselen at different concentrations (0, 1, 5, 10 µM) was added, alkaline phosphatase (ALP) activity, mineralization and osteogenic-related protein levels were evaluated and an optimum concentration of ebselen was selected. Subsequently, intracellular reactive oxygen species (ROS) generation and the role of the PI3K/AKT pathway were also investigated. RESULTS: Ebselen within a proper range could promote the proliferation of BMSCs. H2O2-induced oxidative stress suppressed osteogenic differentiation of BMSCs, which was verified by the decrease in ALP activity, calcium deposition, Runx2 and ß-catenin expression. However, ebselen could alleviate osteogenic dysfunction of BMSCs. We also observed that ebselen reduced ROS accumulation in H2O2-pretreated BMSCs. Moreover, the pro-osteogenic effects afforded by ebselen were almost abolished by the Akt inhibitor. CONCLUSION: We concluded that ebselen could attenuate osteogenic dysfunction of BMSCs induced by H2O2 through an antioxidant effect and the activation of the PI3K/Akt pathway, suggesting that ebselen has a potential therapeutic effect for patients with metabolic bone diseases.


Assuntos
Antioxidantes/farmacologia , Azóis/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Compostos Organosselênicos/farmacologia , Osteogênese/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Antioxidantes/química , Azóis/química , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Peróxido de Hidrogênio/antagonistas & inibidores , Peróxido de Hidrogênio/farmacologia , Isoindóis , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Compostos Organosselênicos/química , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA