Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
ACS Biomater Sci Eng ; 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38885017

RESUMEN

Osteoarthritis (OA) is a chronic joint disease characterized by cartilage imbalance and disruption of cartilage extracellular matrix secretion. Identifying key genes that regulate cartilage differentiation and developing effective therapeutic strategies to restore their expression is crucial. In a previous study, we observed a significant correlation between the expression of the gene encoding casein kinase-2 interacting protein-1 (CKIP-1) in the cartilage of OA patients and OA severity scores, suggesting its potential involvement in OA development. To test this hypothesis, we synthesized a chondrocyte affinity plasmid, liposomes CKIP-1, to enhance CKIP-1 expression in chondrocytes. Our results demonstrated that injection of CAP-Lipos-CKIP-1 plasmid significantly improved OA joint destruction and restored joint motor function by enhancing cartilage extracellular matrix (ECM) secretion. Histological and cytological analyses confirmed that CKIP-1 maintains altered the phosphorylation of the signal transduction molecule SMAD2/3 of the transforming growth factor-ß (TGF-ß) pathway by promoting the phosphorylation of the 8T, 416S sit. Taken together, this work highlights a novel approach for the precise modulation of chondrocyte phenotype from an inflammatory to a noninflammatory state for the treatment of OA and may be broadly applicable to patients suffering from other arthritic diseases.

2.
ACS Biomater Sci Eng ; 10(6): 3868-3882, 2024 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-38703236

RESUMEN

The reconstruction of bone defects has been associated with severe challenges worldwide. Nowadays, bone marrow mesenchymal stem cell (BMSC)-based cell sheets have rendered this approach a promising way to facilitate osteogenic regeneration in vivo. Extracellular vesicles (EVs) play an essential role in intercellular communication and execution of various biological functions and are often employed as an ideal natural endogenous nanomedicine for restoring the structure and functions of damaged tissues. The perception of polymorphonuclear leukocytes (neutrophils, PMNs) as indiscriminate killer cells is gradually changing, with new evidence suggesting a role for these cells in tissue repair and regeneration, particularly in the context of bone healing. However, the role of EVs derived from PMNs (PMN-EVs) in bone regeneration remains largely unknown, with limited research being conducted on this aspect. In the current study, we investigated the effects of PMN-EVs on BMSCs and the underlying molecular mechanisms as well as the potential application of PMN-EVs in bone regeneration. Toward this end, BMSC-based cell sheets with integrated PMN-EVs (BS@PMN-EVs) were developed for bone defect regeneration. PMN-EVs were found to significantly enhance the proliferation and osteogenic differentiation of BMSCs in vitro. Furthermore, BS@PMN-EVs were found to significantly accelerate bone regeneration in vivo by enhancing the maturation of the newly formed bone in rat calvarial defects; this is likely attributable to the effect of PMN-EVs in promoting the expression of key osteogenic proteins such as SOD2 and GJA1 in BMSCs. In conclusion, our findings demonstrate the crucial role of PMN-EVs in promoting the osteogenic differentiation of BMSCs during bone regeneration. Furthermore, this study proposes a novel strategy for enhancing bone repair and regeneration via the integration of PMN-EVs with BMSC-based cell sheets.


Asunto(s)
Regeneración Ósea , Diferenciación Celular , Vesículas Extracelulares , Células Madre Mesenquimatosas , Neutrófilos , Osteogénesis , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/fisiología , Vesículas Extracelulares/trasplante , Regeneración Ósea/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/fisiología , Animales , Neutrófilos/metabolismo , Neutrófilos/fisiología , Neutrófilos/citología , Ratas , Ratas Sprague-Dawley , Masculino , Proliferación Celular , Humanos
3.
Biomater Sci ; 11(7): 2590-2602, 2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-36804554

RESUMEN

Tumor recurrence caused by metastasis is a major cause of death for patients. Thus, a strategy to manipulate the circulating tumor cells (CTCs, initiators of tumor metastasis ) and eliminate them along with the primary tumor has significant clinical significance for malignant tumor therapy. In this study, a magnet-NIR-pH multi-responsive nanosheet (Fe3O4@SiO2-GO-PEG-FA/AMP-DOX, FGPFAD) was fabricated to capture CTCs in circulation, then magnetically transport them to the primary tumor, and finally perform NIR-dependent photothermal therapy as well as acidic-environment-triggered chemotherapy to destroy both the CTCs and the primary tumor. The FGPFAD nanosheet consists of silica-coated ferroferric oxide nanoparticles (Fe3O4@SiO2, magnetic targeting agent), graphene oxide (GO, photothermal therapy agent), polyethylene glycol (PEG, antifouling agent for sustained circulation), folic acid (FA, capturer of CTCs) and antimicrobial-peptide-conjugated doxorubicin (AMP-DOX, agent for chemotherapy), in which the AMP-DOX was bound to the FGPFAD nanosheet via a cleavable Schiff base to achieve acidic-environment-triggered drug release for tumor-specific chemotherapy. Both in vitro and in vivo results indicated that the effective capture and magnetically guided transfer of CTCs to the primary tumor, as well as the multimodal tumor extermination performed by our FGPFAD nanosheet, significantly inhibited the primary tumor and its metastasis.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Células Neoplásicas Circulantes , Humanos , Dióxido de Silicio , Doxorrubicina/farmacología , Fototerapia/métodos , Polietilenglicoles , Línea Celular Tumoral
4.
Natl Sci Rev ; 9(4): nwac037, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35419207

RESUMEN

Cartilage injuries are often devastating and most cannot be cured because of the intrinsically low regenerative capacity of cartilage tissues. Although stem-cell therapy has shown enormous potential for cartilage repair, the therapeutic outcome has been restricted by low survival rates and poor chondrocyte differentiation in vivo. Here, we report an injectable hybrid inorganic (IHI) nanoscaffold that facilitates fast assembly, enhances survival and regulates chondrogenic differentiation of stem cells. IHI nanoscaffolds that strongly bind to extracellular matrix (ECM) proteins assemble stem cells through synergistic 3D cell-cell and cell-matrix interactions, creating a favorable physical microenvironment for stem-cell survival and differentiation in vitro and in vivo. Additionally, chondrogenic factors can be loaded into nanoscaffolds with a high capacity, which allows deep, homogenous drug delivery into assembled 3D stem-cell-derived tissues for effective control over the soluble microenvironment of stem cells. The developed IHI nanoscaffolds that assemble with stem cells are injectable. They also scavenge reactive oxygen species and timely biodegrade for proper integration into injured cartilage tissues. Implantation of stem-cell-assembled IHI nanoscaffolds into injured cartilage results in accelerated tissue regeneration and functional recovery. By establishing our IHI nanoscaffold-templated 3D stem-cell assembly method, we provide a promising approach to better overcoming the inhibitory microenvironment associated with cartilage injuries and to advance current stem-cell-based tissue engineering.

5.
Biochem Biophys Res Commun ; 601: 86-92, 2022 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-35231656

RESUMEN

RNA interference (RNAi)-based gene therapy that promotes anabolic bone formation is an effective approach for addressing osteoporosis. However, the selection of target gene and tissue-specific delivery systems has hindered the progression of this strategy. In this study, we identified casein kinase-2 interacting protein-1 encoding gene (Ckip-1), a negative regulator of bone formation, as an effective target of small interfering RNAs (siRNAs) for improving bone mass. Moreover, an impressive (DSS)6-Liposome (Lipos) nanoparticle system that could target the bone formation surface was synthesized to enhance the delivery of Ckip-1 siRNA to osteogenic lineage cells. The in vitro results confirmed that the (DSS)6-Lipos system could efficaciously improve the intracellular delivery of Ckip-1 siRNA without obvious cell toxicity. The in vivo application of the delivery system showed specific accumulation of siRNA in osteogenic cells located around the bone formation surface. Bone-related analysis indicated increased bone mass and improved bone microarchitecture in mice with ovariectomy-induced osteoporosis. Moreover, the biomechanical characteristics of the tibia were enhanced significantly, indicating increased resistance to fragile fracture induced by osteoporosis. Thus, (DSS)6-Lipos-Ckip-1 siRNA-based osteoanabolic therapy may be a promising option for the treatment of osteoporosis.


Asunto(s)
Osteogénesis , Osteoporosis , Animales , Proteínas Portadoras/metabolismo , Femenino , Liposomas , Ratones , Osteogénesis/genética , Osteoporosis/genética , Osteoporosis/metabolismo , Osteoporosis/terapia , Interferencia de ARN , ARN Interferente Pequeño/genética , Tratamiento con ARN de Interferencia
6.
Adv Sci (Weinh) ; 8(19): e2100584, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34382372

RESUMEN

The role of neutrophils in bone regeneration remains elusive. In this study, it is shown that intramuscular implantation of interleukin-8 (IL-8) (commonly recognized as a chemotactic cytokine for neutrophils) at different levels lead to outcomes resembling those of fracture hematoma at various stages. Ectopic endochondral ossification is induced by certain levels of IL-8, during which neutrophils are recruited to the implanted site and are N2-polarized, which then secrete stromal cell-derived factor-1α (SDF-1α) for bone mesenchymal stem cell (BMSC) chemotaxis via the SDF-1/CXCR4 (C-X-C motif chemokine receptor 4) axis and its downstream phosphatidylinositol 3'-kinase (PI3K)/Akt pathway and ß-catenin-mediated migration. Neutrophils are pivotal for recruiting and orchestrating innate and adaptive immunocytes, as well as BMSCs at the initial stage of bone healing and regeneration. The results in this study delineate the mechanism of neutrophil-initiated bone regeneration and interaction between neutrophils and BMSCs, and innate and adaptive immunities. This work lays the foundation for research in the fields of bone regenerative therapy and biomaterial development, and might inspire further research into novel therapeutic options.


Asunto(s)
Regeneración Ósea/fisiología , Fracturas Óseas/metabolismo , Fracturas Óseas/terapia , Interleucina-8/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neutrófilos/metabolismo , Animales , Huesos/metabolismo , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología
7.
Acta Biomater ; 130: 435-446, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34089908

RESUMEN

Metabolic skeletal disorders remain a major clinical challenge. The complexity of this disease requires a strategy to address the net effects of both inflammation and impaired bone formation. microRNA-based gene therapy provides several therapeutic advantages to tackle these issues. Herein, we describe a microRNA-21 (miR-21) delivery system with an additional therapeutic effect from that of the delivery carrier itself. Poly (salicylic acid) (PSA) is, for the first time, synthesized via polycondensation of salicylic acid (SA), a bioactive ingredient widely used for anti-inflammation in medicine. PSA can self-assemble into nanoparticles (PSA-NPs) and can effectively deliver genes both in vitro and in vivo. The carrier was then attached to repetitive sequences of aspartate, serine, serine (DSS)6 for delivering miRNAs specifically to bone-formation surfaces. In vitro studies showed that miR-21@PSA-NP could effectively realize the intracellular delivery of miR-21 with low toxicity, while in vivo results indicated that the miR-21@PSA-NP-DSS6 prolonged blood circulation time, enhanced bone accumulation, and significantly improved the efficacy of miR-21-based bone anabolic therapy in osteoporotic mice. The constructed delivery system (miR-21@PSA-NP-DSS6) inherited the advantages of both SA and miR-21, which could ameliorate bone-inflamed niche and rescued the impaired bone formation ability. The synergy of anti-inflammatory and pro-osteogenic effects significantly improved trabecular bone microstructure in osteoporotic mice. STATEMENT OF SIGNIFICANCE: The complexity of metabolic skeletal disorders requires a strategy to address the net effects of both inflammation and impaired bone formation. microRNA-based gene therapy provides several therapeutic advantages to tackle these issues. We develop a novel microRNA-21 delivery system with additional therapeutic effect from that of the gene carrier itself. Poly (salicylic acid) (PSA) nanoparticles, for the first time, synthesized via polycondensation of salicylic acid and can effectively deliver genes both in vitro and in vivo. The constructed delivery system (miR-21@PSA-NP-DSS6) inherited the advantages of both SA (commonly used anti-inflammation drug in medicine) and miR-21 (a pro-osteogenic molecule), which could ameliorate bone-inflamed niche, rescued impaired bone formation ability and significantly improved trabecular bone microstructure in osteoporotic mice.


Asunto(s)
MicroARNs , Nanopartículas , Animales , Ratones , MicroARNs/genética , Nanomedicina , Osteogénesis , Ácido Salicílico/farmacología
8.
Biomaterials ; 272: 120718, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33838528

RESUMEN

Mesenchymal stem cell-derived exosomes (MSC-exos), with its inherent capacity to modulate cellular behavior, are emerging as a novel cell-free therapy for bone regeneration. Herein, focusing on practical applying problems, the osteoinductivity of MSC-exos produced by different stem cell sources (rBMSCs/rASCs) and culture conditions (osteoinductive/common) were systematically compared to screen out an optimized osteogenic exosome (BMSC-OI-exo). Via bioinformatic analyses by miRNA microarray and in vitro pathway verification by gene silencing and miRNA transfection, we first revealed that the osteoinductivity of BMSC-OI-exo was attributed to multi-component exosomal miRNAs (let-7a-5p, let-7c-5p, miR-328a-5p and miR-31a-5p). These miRNAs targeted Acvr2b/Acvr1 and regulated the competitive balance of Bmpr2/Acvr2b toward Bmpr-elicited Smad1/5/9 phosphorylation. On these bases, lyophilized delivery of BMSC-OI-exo on hierarchical mesoporous bioactive glass (MBG) scaffold was developed to realize bioactivity maintenance and sustained release by entrapment in the surface microporosity of the scaffold. In a rat cranial defect model, the loading of BMSC-OI-exo efficiently enhanced the bone forming capacity of the scaffold and induced rapid initiation of bone regeneration. This paper could provide empirical bases of MSC-exo-based therapy for bone regeneration and theoretical bases of MSC-exo-induced osteogenesis mechanism. The BMSC-OI-exo-loaded MBG scaffold developed here represented a promising bone repairing strategy for future clinical application.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , MicroARNs , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II , Regeneración Ósea , Liofilización , Osteogénesis , Ratas
9.
Acta Biomater ; 128: 408-419, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33878477

RESUMEN

Gene therapy based on transfection of RNAs/DNAs offers tremendous promise for tumor treatment. However, the relatively weak therapeutic efficiency of current genetic nanohybrids in vivo has limited the application of this strategy. Herein, we fabricated multifunctional core-shell-corona nanohybrids by combining cascaded theranostics for enhanced gene therapy. The nanohybrids consist of polydopamine-modified Fe3O4 nanoparticles as core, anti-miRNA-21 oligonucleotides (anti-miRNA) strands as shell, and doxorubicin (DOX)-conjugated DNA-8pb (DOX-DNA-8bp) as corona. The polydopamine/Fe3O4 core not only serves as an active agent for local photothermal therapy under NIR irradiation, but it also provides magnetic targeting to tumor tissue for accurate treatment, which could enhance the therapeutic effect and reduce the undesired side effects to healthy tissues. The DOX-DNA-8bp corona was grafted on the anti-miRNA shell through base pairing, which could be replaced by overexpressed miRNA-21 in tumor cells due to the strong interaction between miRNA-21 and anti-miRNA, resulting in tumor-specific gene therapy through tumorigenic miRNA-21 consumption and tumor selective chemotherapy through miRNA-21-triggered DOX-DNA-8bp release in tumor cells. Moreover, the intelligent controlled release system can gradually stop the release of DOX to prevent side effects caused by drug overdose, once sufficient damage of tumor cells has occurred, due to the downregulation of miRNA-21. The results of both in vitro and in vivo analyses showed that the nanohybrids combining cascaded chemo-photo-gene therapy could effectively inhibit tumor growth, promote the survival of tumor-bearing mice, and show no visible adverse effects on these mice, resulting in a promising nanoplatform for tumor treatment. STATEMENT OF SIGNIFICANCE: Gene therapy based on transfection of RNAs/DNAs offers tremendous promise for cancer treatment. However, the relatively weak therapeutic efficiency of current genetic nanovectors in vivo that results in insufficient tumor targeting and easy decomposition/elimination of RNAs/DNAs during therapy has limited its application. Although some approaches have combined photothermal agents or antitumor drugs with RNA/DNA nanocarriers to achieve better treatment, the spatiotemporal differences in photothermal therapy, chemotherapy, and gene therapy using current nanohybrids may hinder their synergistic effect. In the present study, we fabricated multifunctional core-shell-corona nanohybrids (Fe3O4@PDA@anti-miRNA/DNA) to simultaneously perform on-demand photothermal therapy, miR-21-triggered chemotherapy, and miR-21-dependent gene therapy at the same location, which can achieve an efficient synergistic effect for precise and effective tumor treatment.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Neoplasias , Animales , Doxorrubicina/farmacología , Ratones , Fototerapia , Medicina de Precisión
10.
Sci Adv ; 7(9)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33627421

RESUMEN

Current therapeutic strategies such as angiogenic therapy and anti-inflammatory therapy for treating myocardial infarction have limited success. An effective approach may benefit from resolution of excessive inflammation combined with enhancement of angiogenesis. Here, we developed a microRNA-21-5p delivery system using functionalized mesoporous silica nanoparticles (MSNs) with additional intrinsic therapeutic effects. These nanocarriers were encapsulated into an injectable hydrogel matrix (Gel@MSN/miR-21-5p) to enable controlled on-demand microRNA-21 delivery triggered by the local acidic microenvironment. In a porcine model of myocardial infarction, we demonstrated that the released MSN complexes notably inhibited the inflammatory response by inhibiting the polarization of M1 macrophage within the infarcted myocardium, while further microRNA-21-5p delivery by MSNs to endothelial cells markedly promoted local neovascularization and rescued at-risk cardiomyocytes. The synergy of anti-inflammatory and proangiogenic effects effectively reduced infarct size in a porcine model of myocardial infarction.


Asunto(s)
MicroARNs , Infarto del Miocardio , Nanopartículas , Animales , Células Endoteliales , Hidrogeles , MicroARNs/genética , Infarto del Miocardio/terapia , Neovascularización Patológica , Dióxido de Silicio , Porcinos
12.
Int J Biol Sci ; 16(16): 3100-3115, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33162818

RESUMEN

Background: Metastasis is the most common cause of lethal outcome in various types of cancers. Although the cell proliferation related metabolism rewiring has been well characterized, less is known about the association of metabolic changes with tumor metastasis. Herein, we demonstrate that metastatic tumor obtained a mesenchymal phenotype, which is obtained by the loss of tumor suppressor NDRG2 triggered metabolic switch to glutamine metabolism. Methods: mRNA-seq and gene expression profile analysis were performed to define the differential gene expressions in primary MEC1 and metastatic MC3 cells and the downstream pathways of NDRG2. NDRG2 regulation of Fbw7-dependent c-Myc stability were determined by immunoprecipitation and protein half-life assay. Luciferase reporter and ChIP assays were used to determine the roles of Akt and c-Myc in mediating NDRG2-dependent regulation of ASCT2 in in both tumor and NDRG2-knockout MEF cells. Finally, the effect of the NDRG2/Akt/c-Myc/ASCT2 signaling on glutaminolysis and tumor metastasis were evaluated by functional experiments and clinical samples. Results: Based on the gene expression profile analysis, we identified metastatic tumor cells acquired the mesenchymal-like characteristics and displayed the increased dependency on glutamine utilization. Further, the gain of NDRG2 function blocked epithelial-mesenchymal transition (EMT) and glutaminolysis, potentially through suppression of glutamine transporter ASCT2 expression. The ASCT2 restoration reversed NDRG2 inhibitory effect on EMT program and tumor metastasis. Mechanistic study indicates that NDRG2 promoted Fbw7-dependent c-Myc degradation by inhibiting Akt activation, and subsequently decreased c-Myc-mediated ASCT2 transcription, in both tumor and NDRG2-knockout MEF cells. Supporting the biological significance, the reciprocal relationship between NDRG2 and ASCT2 were observed in multiple types of tumor tissues, and associated with tumor malignancy. Conclusions: NDRG2-dependent repression of ASCT2 presumably is the predominant route by which NDRG2 rewires glutaminolysis and blocks metastatic tumor survival. Targeting glutaminolytic pathway may provide a new strategy for the treatment of metastatic tumors.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/genética , Reprogramación Celular , Glutamina/metabolismo , Antígenos de Histocompatibilidad Menor/genética , Neoplasias Experimentales/genética , Proteínas Supresoras de Tumor/genética , Animales , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Desnudos , Metástasis de la Neoplasia/genética , Neoplasias Experimentales/metabolismo , Transducción de Señal
13.
Biomaterials ; 253: 120095, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32445809

RESUMEN

Defects of either articular cartilage or subchondral bone would destroy the structural integrity and functionality of the joint. Reconstruction of osteochondral defects requires difunctional scaffolds that simultaneously induce cartilage and subchondral bone morphogenesis, however, high-performance cartilage reconstructive scaffolds remain a considerable challenge. In this study, a solvent-free urethane crosslinking and spontaneous pore-forming procedure under room temperature was proposed and optimized to produce PEGylated poly(glycerol sebacate) (PEGS) scaffolds with controllable crosslinking degrees and hierarchical macro-/micro-porosities. Based on the economical and feasible preparative approach, the viscoelastic PEGS-12h with low crosslinking degree was demonstrated to significantly stimulate chondrogenic differentiation, maintain chondrocyte phenotype and enhance cartilage matrix secretion compared to elastic polymer with high crosslinking degree, emphasizing the importance of matrix viscoelasticity in cartilage regeneration. On this basis, the viscoelastic low-crosslinked PEGS-12h was combined with the well-acknowledged osteoinductive mesoporous bioactive glass (MBG) to construct a difunctional PEGS/MBG bilayer scaffold, and evaluated in a full-thickness osteochondral defect model in vivo. The PEGS/MBG bilayer scaffold successfully reconstructed well-integrated articular hyaline cartilage and its subchondral bone in 12 weeks, exhibiting extraordinary regenerative efficiency. The results indicated that the viscoelastic PEGS scaffold and PEGS/MBG bilayer scaffold proposed in this study made an excellent candidate for cartilage and osteochondral regeneration, and was expected for clinical translation in the future.


Asunto(s)
Cartílago Articular , Andamios del Tejido , Decanoatos , Glicerol/análogos & derivados , Polietilenglicoles , Polímeros , Ingeniería de Tejidos
14.
Biomater Sci ; 8(7): 1865-1874, 2020 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-32021991

RESUMEN

Herein, a rambutan-like nanocomplex (PDA-SNO-GA-HA-DOX, PSGHD for short) was designed to enable effective and accurate tumor therapy. The PSGHD nanocomplex consists of an S-nitrosothiol-functionalized polydopamine (PDA-SNO) core and a gambogic acid-derivatized hyaluronic acid (HA-GA) shell with doxorubicin (DOX) as the cargo. Due to the HA section, the PSGHD nanocomplex can be rapidly and selectively internalized by tumor cells instead of healthy cells in 12 h of co-incubation. After that, the internalized PSGHD nanocomplex is able to gradually release both DOX (agent for chemotherapy) and GA (agent for enhancing thermal damage) under different tumor-specific physiological conditions (low pH and rich HAase). When 808 nm NIR radiation was employed, the PSGHD nanocomplex further demonstrated excellent photothermal conversion to increase the local temperature over 43 °C and convert SNO to nitric oxide (NO, an agent for decreasing drug-efflux). Based on the synergistic effects of NO/DOX and GA/heat, the PSGHD nanocomplex simultaneously achieved tumor-specific low-drug-efflux chemotherapy and low-temperature photothermal therapy, resulting in an eight-fold apoptosis of tumor cells over normal cells under NIR radiation. In vivo data from mouse models further showed that the PSGHD nanocomplex can completely inhibit tumor growth and significantly prolong the survival rate of tumor bearing mice in 50 days, demonstrating the high efficiency of the PSGHD nanocomplex for tumor therapy.


Asunto(s)
Carcinoma de Células Escamosas/cirugía , Doxorrubicina/administración & dosificación , Ácido Hialurónico/administración & dosificación , Neoplasias de la Lengua/terapia , Xantonas/química , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Terapia Combinada , Doxorrubicina/química , Doxorrubicina/farmacología , Humanos , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Hipertermia Inducida , Ratones , Nanocompuestos/química , Fototerapia
15.
J Mater Chem B ; 8(31): 6857-6865, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32022092

RESUMEN

Catalytic medicine based on various catalysts has attracted increasing interest for the treatment of tumors. However, the direct application of conventional catalysts may cause serious side effects to healthy tissue and low therapeutic efficiency against tumor tissue, due to their weak specificity for the tumor microenvironment (TME). Herein, a tumor-targeting and TME-responsive nanoreactor containing ferroferric oxide nanoparticles (Fe3O4 NPs) and glucose oxidase (GOD) was developed to perform hyaluronidase (HAase) and glutathione (GSH)-triggered chain catalytic reactions in tumor tissue. This nanoreactor was designed to take advantage of the unique biological molecules in tumors and several therapeutic agents to adjust the local microenvironments, and achieved satisfactory and accurate tumor therapy. The reactions started with the consumption of intratumoral glucose to inhibit tumor growth, and simultaneously produced hydrogen peroxide (H2O2) to make up for the deficiency of H2O2 in the original tumor microenvironment, resulting in the generation of a high quantity of hydroxyl radicals as a result of the catalysis of Fe3O4 NPs to further eliminate tumor tissue. The tumor-specific catalytic medicine formed by our nanocomposite guaranteed both therapeutic efficiency and accuracy, avoiding potential risks to healthy tissue and leading to a four-fold improvement in the cytotoxicity against tumor cells compared with normal cells after incubations of 48 h. In vivo data from mouse models provided further evidence for its therapeutic efficacy: the tumor growth was completely inhibited after two weeks of the synergistic therapy, which indicated the promise of our nanocomposite for tumor treatment.


Asunto(s)
Biocatálisis , Nanomedicina/métodos , Animales , Línea Celular Tumoral , Proliferación Celular , Glucosa Oxidasa/metabolismo , Glutatión/metabolismo , Humanos , Hialuronoglucosaminidasa/metabolismo , Radical Hidroxilo/metabolismo , Nanopartículas Magnéticas de Óxido de Hierro/química , Ratones , Microambiente Tumoral
16.
ACS Appl Mater Interfaces ; 11(32): 28621-28630, 2019 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-31293148

RESUMEN

Multiple amino acid (glutamine and lysine)-modified gold nanoparticles a with pH-switchable zwitterionic surface were fabricated through coordination bonds using ferrous iron (Fe2+) as bridge ions, which are able to spontaneously and selectively assemble in tumor cells for accurate tumor therapy combining enzyme-triggered photothermal therapy and H2O2-dependent catalytic medicine. These gold nanoparticles showed electric neutrality at pH 7.4 (hematological system) to prevent endocytosis of normal cells, which could be positively charged at pH 6.8 (tumor microenvironment) to promote the endocytosis of tumor cells to these nanoparticles, performing great tumor selectivity. After cell uptake, the specific enzyme (transglutaminase) in tumor cells would catalyze the polymerization of glutamine and lysine to cause the intracellular assembly of these gold nanoparticles, resulting in an excellent photothermal property for accurate tumor therapy. Moreover, the Fe2+ ion could decompose excess hydrogen peroxide (H2O2) in tumor cells via the Fenton reaction, resulting in a large amount of hydroxyl radicals (·OH). These radicals would also cause tumor cell damage. This synergetic therapy associating with high tumor selectivity generated an 8-fold in vitro cytotoxicity against tumor cells compared with normal cells under 48 h incubation with 10 min NIR irradiation. Moreover, in vivo data from tumor-bearing nude mice models showed that tumors can be completely inhibited and gradually eliminated after multimode treatment combining catalytic medicine and photothermal therapy for 3 weeks. This system takes advantage of three tumor microenvironment conditions (low pH, enzyme, and H2O2) to trigger the therapeutic actions, which is a promising platform for cancer therapy that achieved prolonged circulation time in the blood system, selective cellular uptake, and accurate tumor therapy in multiple models.


Asunto(s)
Oro , Hipertermia Inducida , Melanoma Experimental , Nanopartículas del Metal , Proteínas de Neoplasias/metabolismo , Fototerapia , Transglutaminasas/metabolismo , Aminoácidos/química , Aminoácidos/farmacocinética , Aminoácidos/farmacología , Animales , Línea Celular Tumoral , Materiales Biocompatibles Revestidos , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Endocitosis/efectos de los fármacos , Femenino , Oro/química , Oro/farmacocinética , Oro/farmacología , Humanos , Melanoma Experimental/enzimología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Am J Med Sci ; 358(3): 235-242, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31324362

RESUMEN

BACKGROUND: This study aimed to investigate whether glutaminase (GLS) and glutamine synthetase (GS) are involved in c-Myc-mediated tumor development in oral cancer. METHODS: The correlation between the expressions of c-Myc, GLS, and GS in clinical samples and the clinicopathologic features of oral cancer were examined using immunohistochemistry and quantitative real-time polymerase chain reaction. After overexpressing the c-Myc gene and using an inhibitor of GLS or GS, functional experiments were performed to confirm the effects of c-Myc, GLS and GS on proliferation, cell cycle and migration in KB oral cancer cells. The expressions of E-cadherin and N-cadherin were determined by immunofluorescence assays in KB cells overexpressing c-Myc in the presence of GLS or GS inhibitors. RESULTS: The protein expression of GS was correlated with the Tumor, Lymph Node, and Metastasis (TNM) stage. In addition, c-Myc mRNA levels were positively correlated with GS mRNA levels. Overexpression of c-Myc increased the colonies derived from oral cancer cells and caused more cells to be in S phase compared with the mock-vehicle group. The migratory speed of KB cells was promoted by overexpression of c-Myc compared to the mock-vehicle group. However, these effects were effectively reversed in the presence of GLS or GS inhibitor. Furthermore, c-Myc could inhibit E-cadherin protein expression while promoting N-cadherin expression by enhancing the activity of GLS and GS. CONCLUSIONS: c-Myc overexpression promotes oral cancer cell proliferation and migration by enhancing GLS and GS activity. Our findings are beneficial for the identification of novel molecular targets for the prevention and treatment of oral cancer.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular/fisiología , Glutamato-Amoníaco Ligasa/metabolismo , Glutaminasa/metabolismo , Neoplasias de la Boca/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Adulto , Anciano , Femenino , Perfilación de la Expresión Génica , Glutamato-Amoníaco Ligasa/genética , Glutaminasa/genética , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/enzimología , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/fisiología , Adulto Joven
18.
Oncol Rep ; 41(2): 727-741, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30483813

RESUMEN

Epithelial­mesenchymal transition (EMT) is required for the distant metastasis of tumors. The degree of tumor malignancy increases as EMT progresses. Notably, the biology of tumor cells differs from that of normal cells, with regards to characteristics and energy metabolism mechanisms; abnormal glucose metabolism, excessive accumulation of fatty acids and other metabolic disorders occur in metastatic tumors. Previous studies have confirmed that the regulation of tumor cell metabolism can affect tumor metastasis and some findings have resulted in novel clinical applications. The present review aimed to provide a basis for treatments targeting the tumor EMT process and metabolic reprogramming.


Asunto(s)
Antineoplásicos/farmacología , Biomarcadores de Tumor/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Redes y Vías Metabólicas/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Aminoácidos/metabolismo , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/antagonistas & inhibidores , Progresión de la Enfermedad , Metabolismo Energético/efectos de los fármacos , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neoplasias/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Ácidos Nucleicos/metabolismo
19.
J Craniomaxillofac Surg ; 46(12): 2248-2255, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30389311

RESUMEN

PURPOSE: Views on treatment procedures for condylar head fractures (CHFs) are far from reaching a consensus. The aim of this study was to evaluate the changes in disc status for anteromedial disc displacement with anchorage (AMDDwA) and without anchorage (AMDDwoA - just suturing to the adjacent TMJ soft tissue) in adult CHFs, to get a better understanding of this very complex process and to show that rigid disc anchorage is an essential technique for the treatment of CHF during the open reduction and internal fixation (ORIF). PATIENTS AND METHODS: 144 temporomandibular joints (TMJ) in 95 patients were included in this retrospective study, and were divided into an AMDDwA group (50 TMJs in 38 patients) and an AMDDwoA group (94 TMJs in 57 patients) based on the different surgical procedures. The joints were quantitatively and qualitatively assessed for disc length and disc morphology preoperatively and at follow-up visits. Other variables, such as disc position, joint effusion, retrodiscal tear and lateral capsular tear, were also evaluated. Paired t-tests, Wilcoxon signed rank tests, independent t-tests and χ2 tests were used to assess intragroup and intergroup differences. RESULTS: The results showed that discs became shorter, moved further forward and distorted more seriously in the AMDDwoA group. In contrast, discs became longer, maintained a normal disc-condyle relationship in the AMDDwA group. Joint effusion, retrodiscal tear, and lateral capsular tear healed well in both groups. CONCLUSION: Taking these findings together suggests that the rigid disc anchorage is an alternative technique for the treatment of CHF.


Asunto(s)
Fijación Interna de Fracturas/métodos , Cóndilo Mandibular/lesiones , Cóndilo Mandibular/cirugía , Fracturas Mandibulares/cirugía , Disco de la Articulación Temporomandibular/cirugía , Adolescente , Adulto , Anciano , Femenino , Fijación Interna de Fracturas/instrumentación , Humanos , Imagen por Resonancia Magnética , Masculino , Cóndilo Mandibular/diagnóstico por imagen , Fracturas Mandibulares/diagnóstico por imagen , Persona de Mediana Edad , Estudios Retrospectivos , Anclas para Sutura , Técnicas de Sutura , Disco de la Articulación Temporomandibular/diagnóstico por imagen , Resultado del Tratamiento
20.
J Oral Maxillofac Surg ; 76(5): 1026-1035, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29360456

RESUMEN

PURPOSE: In response to the increased attention to soft tissue reduction in the treatment of intracapsular condylar fractures (ICFs), a modified open reduction technique is proposed and its functional and radiographic outcomes were evaluated in this study. PATIENTS AND METHODS: This is a retrospective case series study of patients with all ICF types that were treated with open reduction and internal fixation (ORIF) with articular disc anatomic reduction and rigid anchorage. Inclusion and exclusion criteria were strictly applied. Preoperative and postoperative clinical examinations of malocclusion, maximum incisor opening (MIO), laterotrusion, and temporomandibular disorder symptoms were recorded and analyzed. Computed tomography (CT) and magnetic resonance imaging (MRI) were used to assess articular position and condylar morphology and position. RESULTS: Thirty-four patients with ICFs (47 sides) were treated with the modified ORIF technique. At 6 months of follow-up, no malocclusion was found and the MIO considerably expanded to 3.56 ± 0.13 cm. Only 4 patients (12%) had temporomandibular joint discomfort with mouth opening. Interestingly, for unilateral type B ICFs, the laterotrusion distance to the ORIF sides was notably longer than to the non-ORIF sides. Postoperative CT and MRI showed that all fragments were properly reduced and the condyles were in the normal position. Postoperative anterior disc displacement occurred in 4 sides and condylar morphologic abnormalities (slight surface roughening and articular cartilage absorption) occurred in 3 sides (6.4%). CONCLUSIONS: This modified ORIF technique, which achieved good outcomes after treatment of all ICF types, shows promise for the treatment of ICFs.


Asunto(s)
Fijación Interna de Fracturas/métodos , Cápsula Articular/lesiones , Cóndilo Mandibular/lesiones , Fracturas Mandibulares/cirugía , Reducción Abierta/métodos , Articulación Temporomandibular/lesiones , Adolescente , Adulto , Anciano , Femenino , Estudios de Seguimiento , Humanos , Cápsula Articular/cirugía , Masculino , Cóndilo Mandibular/cirugía , Persona de Mediana Edad , Estudios Retrospectivos , Articulación Temporomandibular/cirugía , Resultado del Tratamiento , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA