Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Mater Chem B ; 11(33): 8007-8019, 2023 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-37530140

RESUMO

Craniofacial/jawbone deformities remain a significant clinical challenge in restoring facial/dental functions and esthetics. Despite the reported therapeutics for clinical bone tissue regeneration, the bioavailability issue of autografts and limited regeneration efficacy of xenografts/synthetic bone substitutes, however, inspire continued efforts towards functional conjugation and improvement of bioactive bone graft materials. Regarding the potential of nitric oxide (NO) in tissue engineering, herein, functional conjugation of NO-delivery dinitrosyl iron complex (DNIC) and osteoconductive bone graft materials was performed to optimize the spatiotemporal control over the delivery of NO and to activate synergistic osteogenesis and angiogenesis in rat calvaria bone defects. Among three types of biomimetic DNICs, [Fe2(µ-SCH2CH2COOH)2(NO)4] (DNIC-COOH) features a steady kinetics for cellular uptake by MC3T3-E1 osteoblast cells followed by intracellular assembly of protein-bound DNICs and release of NO. This steady kinetics for intracellular delivery of NO by DNIC-COOH rationalizes its biocompatibility and wide-spectrum cell proliferation effects on MC3T3-E1 osteoblast cells and human umbilical vein endothelial cells (HUVECs). Moreover, the bridging [SCH2CH2COOH]- thiolate ligands in DNIC-COOH facilitate its chemisorption to deproteinized bovine bone mineral (DBBM) and physisorption onto TCP (ß-tricalcium phosphate), respectively, which provides a mechanism to control the kinetics for the local release of loaded DNIC-COOH. Using rats with calvaria bone defects as an in vivo model, DNIC-DBBM/DNIC-TCP promotes the osteogenic and angiogenic activity ascribed to functional conjugation of osteoconductive bone graft materials and NO-delivery DNIC-COOH. Of importance, the therapeutic efficacy of DNIC-DBBM/DNIC-TCP on enhanced compact bone formation after treatment for 4 and 12 weeks supports the potential for clinical application to regenerative medicine.


Assuntos
Óxido Nítrico , Osteogênese , Ratos , Humanos , Animais , Bovinos , Ferro/farmacologia , Células Endoteliais da Veia Umbilical Humana , Crânio
2.
Bioconjug Chem ; 34(9): 1688-1703, 2023 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-37552618

RESUMO

The employment of metal-organic framework (MOF)-based nanomaterials has been rapidly increasing in bioapplications owing to their biocompatibility, drug degradation, tunable porosity, and intrinsic biodegradability. This evidence suggests that the multifunctional bimetallic ions can behave as remarkable candidates for infection control and wound healing. In this study, bimetallic MOFs (Zn-HKUST-1 and FolA-Zn-HKUST-1) embedded with and without folic acid were synthesized and used for tissue sealing and repairing incisional wound sites in mice models. For comparison, HKUST-1 and FolA-HKUST-1 were also synthesized. The Brunauer-Emmett-Teller (BET) surface area measured for HKUST-1, FolA-HKUST-1, Zn-HKUST-1, and FolA-Zn-HKUST-1 from N2 isotherms was found to be 1868, 1392, 1706, and 1179 m2/g, respectively. The measurements of contact angle values for Zn-HKUST-1, FolA-HKUST-1, and Zn-FolA-HKUST-1 were identified as 4.95 ± 0.8, 43.6 ± 3.4, and 60.62 ± 2.0°, respectively. For topical application in wound healing, they display a wide range of healing characteristics, including antibacterial and enhanced wound healing rates. In addition, in vitro cell migration and tubulogenic potentials were evaluated. The significant reduction in the wound gap and increased expression levels for CD31, eNOS, VEGF-A, and Ki67 were observed from immunohistological analyses to predict the angiogenesis behavior at the incision wound site. The wound healing rate was analyzed in the excisional dermal wounds of diabetic mice model in vivo. On account of antibacterial potentials and tissue-repairing characteristics of Cu2+ and Zn2+ ions, designing an innovative mixed metal ion-based biomaterial has wide applicability and is expected to modulate the growth of various gradient tissues.


Assuntos
Diabetes Mellitus Experimental , Estruturas Metalorgânicas , Camundongos , Animais , Estruturas Metalorgânicas/uso terapêutico , Cobre/uso terapêutico , Diabetes Mellitus Experimental/tratamento farmacológico , Zinco/uso terapêutico , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Bactérias
3.
Biomedicines ; 10(10)2022 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-36289750

RESUMO

The primary cause of breast cancer mortality is the metastatic invasion of cancerous stem cells (CSC). Cluster of differentiation 44 (CD44) is a well-known CSC marker in various cancers, as well as a key role player in metastasis and relapse of breast cancer. CD44 is a cell-membrane embedded protein, and it interacts with different proteins to regulate cancer cell behavior. Transcription factor forkhead box protein A2 (FOXA2) acts as an important regulator in multiple cancers, including breast cancer. However, the biological significance of CD44-FOXA2 association in breast cancer metastasis remains unclear. Herein, we observed that CD44 expression was higher in metastatic lymph nodes compared to primary tumors using a flow cytometric analysis. CD44 overexpression in breast cancer cell lines significantly promoted cell migration and invasion abilities, whereas the opposite effects occurred upon the knockdown of CD44. The stem cell array analysis revealed that FOXA2 expression was upregulated in CD44 knockdown cells. However, the knockdown of FOXA2 in CD44 knockdown cells reversed the effects on cell migration and invasion. Furthermore, we found that CD44 mediated FOXA2 localization in breast cancer cells through the AKT pathway. Moreover, the immunofluorescence assay demonstrated that AKT inhibitor wortmannin and AKT activator SC79 treatment in breast cancer cells impacted FOXA2 localization. Collectively, this study highlights that CD44 promotes breast cancer metastasis by downregulating nuclear FOXA2.

4.
Theranostics ; 8(5): 1435-1448, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29507632

RESUMO

Therapeutic efficacy of glioblastoma multiforme (GBM) is often severely limited by poor penetration of therapeutics through blood-brain barrier (BBB) into brain tissues and lack of tumor targeting. In this regard, a functionalized upconversion nanoparticle (UCNP)-based delivery system which can target brain tumor and convert deep tissue-penetrating near-infrared (NIR) light into visible light for precise phototherapies on brain tumor was developed in this work. Methods: The UCNP-based phototherapy delivery system was acquired by assembly of oleic acid-coated UCNPs with angiopep-2/cholesterol-conjugated poly(ethylene glycol) and the hydrophobic photosensitizers. The hybrid nanoparticles (ANG-IMNPs) were characterized by DLS, TEM, UV/vis and fluorescence spectrophotometer. Cellular uptake was examined by laser scanning confocal microscopy and flow cytometry. The PDT/PTT effect of ANG-IMNPs was evaluated using MTT assay. Tumor accumulation of NPs was determined by a non-invasive in vivo imaging system (IVIS). The in vivo anti-glioma effect of ANG-IMNPs was evaluated by immunohistochemical (IHC) examination of tumor tissues and Kaplan-Meier survival analysis. Results: In vitro data demonstrated enhanced uptake of ANG-IMNPs by murine astrocytoma cells (ALTS1C1) and pronounced cytotoxicity by combined NIR-triggered PDT and PTT. In consistence with the increased penetration of ANG-IMNPs through endothelial monolayer in vitro, the NPs have also shown significantly enhanced accumulation at brain tumor by IVIS. The IHC tissue examination confirmed prominent apoptotic and necrotic effects on tumor cells in mice receiving targeted dual photo-based therapies, which also led to enhanced median survival (24 days) as compared to the NP treatment without angiopep-2 (14 days). Conclusion: In vitro and in vivo data strongly indicate that the ANG-IMNPs were capable of selectively delivering dual photosensitizers to brain astrocytoma tumors for effective PDT/PTT in conjugation with a substantially improved median survival. The therapeutic efficacy of ANG-IMNPs demonstrated in this study suggests their potential in overcoming BBB and establishing an effective treatment against GBM.


Assuntos
Neoplasias Encefálicas/terapia , Sistemas de Liberação de Medicamentos , Glioblastoma/terapia , Hipertermia Induzida , Nanopartículas/uso terapêutico , Fotoquimioterapia , Fototerapia , Animais , Barreira Hematoencefálica/patologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Difusão Dinâmica da Luz , Glioblastoma/patologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , Nanopartículas/química , Nanopartículas/ultraestrutura , Oligopeptídeos/química , Peptídeos Cíclicos , Somatostatina/análogos & derivados , Somatostatina/química , Temperatura , Distribuição Tecidual
5.
ACS Appl Mater Interfaces ; 8(36): 23909-19, 2016 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-27556284

RESUMO

High-yield multibranched Au nanoechinus possessing lengthy and dense branched nanorods on the surface were synthesized using a seed-mediated surfactant-directed approach in the presence of gemini cationic surfactant N,N,N'N'-tetramethyl-N,N'-ditetradecylethane-1,2-diaminium bromide (C14C2C14Br2), HAuCl4, AgNO3, and ascorbic acid. C14C2C14Br2 surfactant provides a versatile template in designing the unique morphology of Au nanoechinus with the assistance of AgNO3. UV-vis spectroscopic analysis proves that Au nanoechinus possess a unique intense broad localized surface plasmon resonance (LSPR) peak, which extends from 400 to 1700 nm in the NIR region making a highly potential platform for biomedical applications. Systematic time-dependent TEM, UV-vis-NIR, and XRD analysis were performed to monitor the morphological evolution of multibranched Au nanoechinus. It was found that the surface of branched nanorods of Au NE preferentially grew along (111) crystal planes. Furthermore, as-synthesized Au nanoechinus shows excellent SERS enhancement ability for dopamine inside HeLa cells.

6.
Biomaterials ; 62: 13-23, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26016691

RESUMO

Cancer is one of the major diseases leading to human deaths. Complete destruction of deep tissue-buried tumors using non-invasive therapies is a grand challenge in clinical cancer treatments. Many therapeutic modalities were developed to tackle this problem, but only partial tumor suppression or delay growths were usually achieved. In this study, we report for the first time that complete destruction of deep tissue-buried tumors can be achieved by combination of gold nanoechinus (Au NEs)-mediated photodynamic therapy (PDT) and gene silencing under ultra-low doses of near infra-red (NIR) light irradiation (915 nm, 340 mW/cm(2); 1064 nm, 420 mW/cm(2)) in the first and second biological windows. The average lifespan of the mice treated by the above combined therapy is beyond 40 days, which are ∼ 2.6 times longer than that (15 days) observed from the anticancer drug doxorubicin-treated group. The current study points out a new direction for the therapeutic design to treat deeply seated tumors in future cancer treatments.


Assuntos
Terapia Genética/métodos , Ouro/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Neoplasias Experimentais/terapia , Fotoquimioterapia/métodos , RNA Interferente Pequeno/administração & dosagem , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Terapia Combinada/métodos , Inativação Gênica , Ouro/química , Células HeLa , Humanos , Nanopartículas Metálicas/química , Camundongos , Nanocápsulas/química , Nanocápsulas/uso terapêutico , Nanocápsulas/ultraestrutura , Neoplasias Experimentais/patologia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/uso terapêutico , RNA Interferente Pequeno/genética , Resultado do Tratamento
7.
Adv Mater ; 26(39): 6689-95, 2014 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-25042520

RESUMO

Gold nanoechinus can sensitize the formation of singlet oxygen in the first and the second near-infra red (NIR) biological windows and exert in vivo dual modal photodynamic and photothermal therapeutic effects (PDT and PTT) to destruct the tumors completely. This is the first literature example of the destruction of tumors in NIR window II induced by dual modal nanomaterial-mediated photodynamic and photothermal therapy (NmPDT & NmPTT).


Assuntos
Desenho de Fármacos , Ouro/química , Ouro/farmacologia , Raios Infravermelhos , Nanoestruturas , Fotoquimioterapia/métodos , Animais , Melanoma Experimental/tratamento farmacológico , Camundongos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA