Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Nanobiotechnology ; 22(1): 213, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38689259

RESUMO

BACKGROUND: The main issues faced during the treatment of apical periodontitis are the management of bacterial infection and the facilitation of the repair of alveolar bone defects to shorten disease duration. Conventional root canal irrigants are limited in their efficacy and are associated with several side effects. This study introduces a synergistic therapy based on nitric oxide (NO) and antimicrobial photodynamic therapy (aPDT) for the treatment of apical periodontitis. RESULTS: This research developed a multifunctional nanoparticle, CGP, utilizing guanidinylated poly (ethylene glycol)-poly (ε-Caprolactone) polymer as a carrier, internally loaded with the photosensitizer chlorin e6. During root canal irrigation, the guanidino groups on the surface of CGP enabled effective biofilm penetration. These groups undergo oxidation by hydrogen peroxide in the aPDT process, triggering the release of NO without hindering the production of singlet oxygen. The generated NO significantly enhanced the antimicrobial capability and biofilm eradication efficacy of aPDT. Furthermore, CGP not only outperforms conventional aPDT in eradicating biofilms but also effectively promotes the repair of alveolar bone defects post-eradication. Importantly, our findings reveal that CGP exhibits significantly higher biosafety compared to sodium hypochlorite, alongside superior therapeutic efficacy in a rat model of apical periodontitis. CONCLUSIONS: This study demonstrates that CGP, an effective root irrigation system based on aPDT and NO, has a promising application in root canal therapy.


Assuntos
Biofilmes , Nanopartículas , Óxido Nítrico , Fotoquimioterapia , Animais , Fotoquimioterapia/métodos , Óxido Nítrico/farmacologia , Óxido Nítrico/metabolismo , Biofilmes/efeitos dos fármacos , Ratos , Nanopartículas/química , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/química , Periodontite Periapical/terapia , Periodontite Periapical/tratamento farmacológico , Masculino , Irrigantes do Canal Radicular/farmacologia , Irrigantes do Canal Radicular/química , Ratos Sprague-Dawley , Infecções Bacterianas/tratamento farmacológico , Clorofilídeos , Antibacterianos/farmacologia , Antibacterianos/química , Anti-Infecciosos/farmacologia , Anti-Infecciosos/química
2.
J Nanobiotechnology ; 19(1): 418, 2021 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-34903226

RESUMO

BACKGROUNDS: Sonodynamic therapy (SDT) as an emerging reactive oxygen species (ROS)-mediated antitumor strategy is challenged by the rapid depletion of oxygen, as well as the hypoxic tumor microenvironment. Instead of the presently available coping strategies that amplify the endogenous O2 level, we have proposed a biodegradable O2 economizer to reduce expenditure for augmenting SDT efficacy in the present study. RESULTS: We successfully fabricated the O2 economizer (HMME@HMONs-3BP-PEG, HHBP) via conjugation of respiration inhibitor 3-bromopyruvate (3BP) with hollow mesoporous organosilica nanoparticles (HMONs), followed by the loading of organic sonosensitizers (hematoporphyrin monomethyl ether; HMME) and further surface modification of poly(ethylene glycol) (PEG). The engineered HHBP features controllable pH/GSH/US-sensitive drug release. The exposed 3BP could effectively inhibit cell respiration for restraining the oxygen consumption, which could alleviate the tumor hypoxia conditions. More interestingly, it could exorbitantly elevate the autophagy level, which in turn induced excessive activation of autophagy for promoting the therapeutic efficacy. As a result, when accompanied with suppressing O2-consumption and triggering pro-death autophagy strategy, the HHBP could achieve the remarkable antitumor activity, which was systematically validated both in vivo and in vitro assays. CONCLUSIONS: This work not only provides a reduce expenditure means for enduring SDT, but also represents an inquisitive strategy for tumor treatments by inducing pro-death autophagy.


Assuntos
Autofagia/efeitos dos fármacos , Nanopartículas/química , Compostos de Organossilício/química , Hipóxia Tumoral/efeitos dos fármacos , Terapia por Ultrassom , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Reatores Biológicos , Feminino , Hematoporfirinas , Camundongos , Camundongos Nus , Polietilenoglicóis , Espécies Reativas de Oxigênio/química
3.
J Nanobiotechnology ; 19(1): 345, 2021 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-34715854

RESUMO

BACKGROUND: The risk of local recurrence after high-intensity focused ultrasound (HIFU) is relatively high, resulting in poor prognosis of malignant tumors. The combination of HIFU with traditional chemotherapy continues to have an unsatisfactory outcome because of off-site drug uptake. RESULTS: Herein, we propose a strategy of inflammation-tendency neutrophil-mediated clinical nanodrug targeted therapy for residual tumors after HIFU ablation. We selected neutrophils as carriers and PEGylated liposome doxorubicin (PLD) as a model chemotherapeutic nanodrug to form an innovative cell therapy drug (PLD@NEs). The produced PLD@NEs had a loading capacity of approximately 5 µg of PLD per 106 cells and maintained the natural characteristics of neutrophils. The targeting performance and therapeutic potential of PLD@NEs were evaluated using Hepa1-6 cells and a corresponding tumor-bearing mouse model. After HIFU ablation, PLD@NEs were recruited to the tumor site by inflammation (most in 4 h) and released PLD with inflammatory stimuli, leading to targeted and localized postoperative chemotherapy. CONCLUSIONS: This effective integrated method fully leverages the advantages of HIFU, chemotherapy and neutrophils to attract more focus on the practice of improving existing clinical therapies.


Assuntos
Antineoplásicos/farmacologia , Nanomedicina , Neoplasia Residual/tratamento farmacológico , Neutrófilos , Terapia por Ultrassom , Animais , Linhagem Celular Tumoral , Doxorrubicina/análogos & derivados , Ablação por Ultrassom Focalizado de Alta Intensidade/métodos , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Polietilenoglicóis
4.
J Am Chem Soc ; 138(31): 9881-94, 2016 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-27441571

RESUMO

Biodegradability of inorganic nanoparticles is one of the most critical issues in their further clinical translations. In this work, a novel "metal ion-doping" approach has been developed to endow inorganic mesoporous silica-based nanoparticles with tumor-sensitive biodegradation and theranostic functions, simply by topological transformation of mesoporous silica to metal-doped composite nanoformulations. "Manganese extraction" sensitive to tumor microenvironment was enabled in manganese-doped hollow mesoporous silica nanoparticles (designated as Mn-HMSNs) to fast promote the disintegration and biodegradation of Mn-HMSNs, further accelerating the breakage of Si-O-Si bonds within the framework. The fast biodegradation of Mn-HMSNs sensitive to mild acidic and reducing microenvironment of tumor resulted in much accelerated anticancer drug releasing and enhanced T1-weighted magnetic resonance imaging of tumor. A high tumor-inhibition effect was simultaneously achieved by anticancer drug delivery mediated by PEGylated Mn-HMSNs, and the high biocompatibility of composite nanosystems was systematically demonstrated in vivo. This is the first demonstration of biodegradable inorganic mesoporous nanosystems with specific biodegradation behavior sensitive to tumor microenvironment, which also provides a feasible approach to realize the on-demand biodegradation of inorganic nanomaterials simply by "metal ion-doping" strategy, paving the way to solve the critical low-biodegradation issue of inorganic drug carriers.


Assuntos
Manganês/química , Nanopartículas/química , Neoplasias/diagnóstico por imagem , Animais , Antineoplásicos/química , Materiais Biocompatíveis/química , Sobrevivência Celular , Doxorrubicina/química , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Feminino , Células Hep G2 , Humanos , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia Eletrônica de Transmissão , Nanoestruturas , Neoplasias/patologia , Oxigênio/química , Silício/química , Dióxido de Silício/química , Nanomedicina Teranóstica , Termodinâmica
5.
Adv Sci (Weinh) ; 11(24): e2308587, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38647388

RESUMO

The treatment of diabetic periodontitis poses a significant challenge due to the presence of local inflammation characterized by excessive glucose concentration, bacterial infection, and high oxidative stress. Herein, mesoporous silica nanoparticles (MSN) are embellished with gold nanoparticles (Au NPs) and loaded with manganese carbonyl to prepare a carbon monoxide (CO) enhanced multienzyme cooperative hybrid nanoplatform (MSN-Au@CO). The Glucose-like oxidase activity of Au NPs catalyzes the oxidation of glucose to hydrogen peroxide (H2O2) and gluconic acid,and then converts H2O2 to hydroxyl radicals (•OH) by peroxidase-like activity to destroy bacteria. Moreover, CO production in response to H2O2, together with Au NPs exhibited a synergistic anti-inflammatory effect in macrophages challenged by lipopolysaccharides. The underlying mechanism can be the induction of nuclear factor erythroid 2-related factor 2 to reduce reactive oxygen species, and inhibition of nuclear factor kappa-B signaling to diminish inflammatory response. Importantly, the antibacterial and anti-inflammation effects of MSN-Au@CO are validated in diabetic rats with ligature-induced periodontitis by showing decreased periodontal bone loss with good biocompatibility. To summarize, MSN-Au@CO is fabricate to utilize glucose-activated cascade reaction to eliminate bacteria, and synergize with gas therapy to regulate the immune microenvironment, offering a potential direction for the treatment of diabetic periodontitis.


Assuntos
Monóxido de Carbono , Diabetes Mellitus Experimental , Ouro , Nanopartículas Metálicas , Periodontite , Animais , Periodontite/metabolismo , Periodontite/tratamento farmacológico , Ouro/química , Ratos , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Monóxido de Carbono/metabolismo , Monóxido de Carbono/farmacologia , Modelos Animais de Doenças , Catálise , Ratos Sprague-Dawley , Masculino
6.
J Colloid Interface Sci ; 663: 787-800, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38442520

RESUMO

Ferroptosis is characterized by the lethal accumulation of lipid reactive oxygen species (ROS), which has great potential for tumor therapy. However, developing new ferroptosis-inducing strategies by combining nanomaterials with small molecule inducers is important. In this study, an enzyme-gated biodegradable natural-product delivery system based on lactate oxidase (LOD)-gated biodegradable iridium (Ir)-doped hollow mesoporous organosilica nanoparticles (HMONs) loaded with honokiol (HNK) (HNK@Ir-HMONs-LOD, HIHL) is designed to enhance ferroptosis in colon tumor therapy. After reaching the tumor microenvironment, the outer LOD dissociates and releases the HNK to induce ferroptosis. Moreover, the released dopant Ir4+ and disulfide-bridged organosilica frameworks deplete intracellular glutathione (GSH), which is followed by GSH-mediated Ir(IV)/Ir(III) conversion. This leads to the repression of glutathione peroxidase 4 (GPX4) activity and decomposition of intratumoral hydrogen peroxide (H2O2) into hydroxyl radicals (•OH) by Ir3+-mediated Fenton-like reactions. Moreover, LOD efficiently depletes lactic acid to facilitate the generation of H2O2 and boost the Fenton reaction, which in turn enhances ROS generation. With the synergistic effects of these cascade reactions and the release of HNK, notable ferroptosis efficacy was observed both in vitro and in vivo. This combination of natural product-induced and lactic acid-responsive sequential production of H2O2 as well as the consumption of glutathione may provide a new paradigm for achieving effective ferroptosis-based cancer therapy.


Assuntos
Compostos Alílicos , Compostos de Bifenilo , Neoplasias do Colo , Ferroptose , Lignanas , Fenóis , Humanos , Peróxido de Hidrogênio , Espécies Reativas de Oxigênio , Glutationa , Materiais Biocompatíveis , Irídio , Ácido Láctico , Linhagem Celular Tumoral , Microambiente Tumoral
7.
Small ; 8(5): 760-9, 2012 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-22228696

RESUMO

In biomedical applications, polyethylene glycol (PEG) functionalization has been a major approach to modify nanocarriers such as nano-graphene oxide for particular biological requirements. However, incorporation of a PEG shell poses a significant diffusion barrier that adversely affects the release of the loaded drugs. This study addresses this critical issue by employing a redox-responsive PEG detachment mechanism. A PEGylated nano-graphene oxide (NGO-SS-mPEG) with redox-responsive detachable PEG shell is developed that can rapidly release an encapsulated payload at tumor-relevant glutathione (GSH) levels. The PEG shell grafted onto NGO sheets gives the nanocomposite high physiological solubility and stability in circulation. It can selectively detach from NGO upon intracellular GSH stimulation. The surface-engineered structures are shown to accelerate the release of doxorubicin hydrochloride (DXR) from NGO-SS-mPEG 1.55 times faster than in the absence of GSH. Confocal microscopy shows clear evidence of NGO-SS-mPEG endocytosis in HeLa cells, mainly accumulated in cytoplasm. Furthermore, upon internalization of DXR-loaded NGO with a disulfide-linked PEG shell into HeLa cells, DXR is effectively released in the presence of an elevated GSH reducing environment, as observed in confocal microscopy and flow cytometric experiments. Importantly, inhibition of cell proliferation is directly correlated with increased intracellular GSH concentrations due to rapid DXR release.


Assuntos
Doxorrubicina/administração & dosagem , Doxorrubicina/química , Portadores de Fármacos/química , Grafite/química , Polietilenoglicóis/química , Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos , Endocitose/efeitos dos fármacos , Citometria de Fluxo , Glutationa/metabolismo , Células HeLa , Humanos , Microscopia Confocal , Oxirredução/efeitos dos fármacos
8.
Biomacromolecules ; 13(4): 1024-34, 2012 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-22443494

RESUMO

A dual stimulus-responsive mPEG-SS-PLL(15)-glutaraldehyde star (mPEG-SS-PLL(15)-star) catiomer is developed and biologically evaluated. The catiomer system combines redox-sensitive removal of an external PEG shell with acid-induced escape from the endosomal compartment. The design rationale for PEG shell removal is to augment intracellular uptake of mPEG-SS-PLL(15)-star/DNA complexes in the presence of tumor-relevant glutathione (GSH) concentration, while the acid-induced dissociation is to accelerate the release of genetic payload following successful internalization into targeted cells. Size alterations of complexes in the presence of 10 mM GSH suggest stimulus-induced shedding of external PEG layers under redox conditions that intracellularly present in the tumor microenvironment. Dynamic laser light scattering experiments under endosomal pH conditions show rapid destabilization of mPEG-SS-PLL(15)-star/DNA complexes that is followed by facilitating efficient release of encapsulated DNA, as demonstrated by agarose gel electrophoresis. Biological efficacy assessment using pEGFP-C1 plasmid DNA encoding green fluorescence protein and pGL-3 plasmid DNA encoding luciferase as reporter genes indicate comparable transfection efficiency of 293T cells of the catiomer with a conventional polyethyleneimine (bPEI-25k)-based gene delivery system. These experimental results show that mPEG-SS-PLL(15)-star represents a promising design for future nonviral gene delivery applications with high DNA binding ability, low cytotoxicity, and high transfection efficiency.


Assuntos
Reagentes de Ligações Cruzadas/química , Dissulfetos/química , Técnicas de Transferência de Genes , Vetores Genéticos/química , Iminas/química , Polilisina/química , Cátions/síntese química , Cátions/química , Cátions/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Reagentes de Ligações Cruzadas/síntese química , Reagentes de Ligações Cruzadas/farmacologia , Dissulfetos/farmacologia , Vetores Genéticos/síntese química , Vetores Genéticos/farmacologia , Glutaral/química , Glutaral/farmacologia , Células HEK293 , Células HeLa , Humanos , Iminas/farmacologia , Estrutura Molecular , Oxirredução , Tamanho da Partícula , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Polilisina/genética , Propriedades de Superfície
9.
Theranostics ; 11(7): 3213-3228, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33537083

RESUMO

Rationale: Acute pancreatitis (AP) is a serious acute condition affecting the abdomen and shows high morbidity and mortality rates. Its global incidence has increased in recent years. Inflammation and oxidative stress are potential therapeutic targets for AP. This study was conducted to investigate the intrinsic anti-oxidative and anti-inflammatory effects of Prussian blue nanozyme (PBzyme) on AP, along with its underlying mechanism. Methods: Prussian blue nanozymes were prepared by polyvinylpyrrolidone modification method. The effect of PBzyme on inhibiting inflammation and scavenging reactive oxygen species was verified at the cellular level. The efficacy and mechanism of PBzyme for prophylactically treating AP were evaluated using the following methods: serum testing in vivo, histological scoring following hematoxylin and eosin staining, terminal deoxynucleotidyl transferase dUTP nick end labeling fluorescence staining, polymerase chain reaction array, Kyoto Encyclopedia of Genes and Genomes analysis and Western blotting analysis. Results: The synthetic PBzyme showed potent anti-oxidative and anti-inflammatory effects in reducing oxidative stress and alleviating inflammation both in vitro and in vivo in the prophylactic treatment of AP. The prophylactic therapeutic efficacy of PBzyme on AP may involve inhibition of the toll-like receptor/nuclear factor-κB signaling pathway and reactive oxygen species scavenging. Conclusion: The single-component, gram-level mass production, stable intrinsic biological activity, biosafety, and good therapeutic efficacy suggest the potential of PBzyme in the preventive treatment of AP. This study provides a foundation for the clinical application of PBzyme.


Assuntos
Terapia Enzimática/métodos , Nanotecnologia/métodos , Pancreatite/terapia , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , China , Citocinas/metabolismo , Enzimas/metabolismo , Enzimas/farmacologia , Ferricianetos/química , Ferricianetos/uso terapêutico , Ferrocianetos/química , Ferrocianetos/uso terapêutico , Humanos , Inflamação/tratamento farmacológico , Inflamação/patologia , Masculino , Camundongos Endogâmicos BALB C , NF-kappa B/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Pancreatite/metabolismo , Povidona/química , Povidona/uso terapêutico , Reação do Azul da Prússia/métodos , Espécies Reativas de Oxigênio/metabolismo , Receptores Toll-Like/efeitos dos fármacos
10.
Carbohydr Polym ; 257: 117636, 2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33541661

RESUMO

Development of multifunctional antibacterial agent with long-lasting antibacterial activity and biofilm ablation performance is significant for the effective treatment of bacterial infections. Here, by utilizing the electrostatic interaction between sulfonated chitosan (SCS) and Ag+ and chitosan (CS), and the sodium borohydride reduction method, a versatile antibacterial agent (AgNPs@CS/SCS) capable of generating silver nanoparticles (AgNPs) in-situ and long-acting slow-release Ag+ was developed. AgNPs@CS/SCS has a good physiological stability and can long-acting slow-release of Ag+ due to the pH-dependent Ag+ release behavior of AgNPs. Noteworthy, AgNPs@CS/SCS can exert both excellent short- and long-term antibacterial and biofilm ablation activity. Importantly, it also exhibits superior antibacterial activity in the treatment of implant infections, accompanied by good biocompatibility. Together, this study suggest that AgNPs@CS/CSC is indeed a versatile antibacterial agent, and is expected to provide an effective treatment modality for implant infections in the clinic settings.


Assuntos
Antibacterianos/farmacologia , Materiais Biocompatíveis/química , Quitosana/química , Sistemas de Liberação de Medicamentos , Nanopartículas Metálicas/química , Nanogéis/química , Prata/química , Biofilmes/crescimento & desenvolvimento , Preparações de Ação Retardada/química , Escherichia coli/efeitos dos fármacos , Concentração de Íons de Hidrogênio , Testes de Sensibilidade Microbiana , Staphylococcus aureus/efeitos dos fármacos , Eletricidade Estática
11.
Biomater Sci ; 8(3): 858-870, 2020 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-31808470

RESUMO

Hypoxic resistance, photosensitizer toxicity, and target deficiency are major challenges strongly inhibiting the efficacy of clinical photodynamic therapy (PDT) in tumor treatment. To overcome these challenges, we synthesized IR780 and catalase co-loaded liposomes to form a tumor-targeted bio-nanoreactor (LIP-IR-CAT). The efficient strategy can solve the physicochemical problems including strong hydrophobicity, poor light stability, poor tolerance, and high toxicity in vivo of IR780 as a photosensitizer and promote the clinical application of IR780. Taking advantage of the high catalytic efficiency of catalase when it meets hydrogen peroxide (H2O2), continuous oxygen can be generated due to the abnormally elevated level of H2O2 within the tumor, thus remarkably promoting tumor oxygenation. With the conjunction of photosensitivity and specific mitochondria-targeting ability of IR780, the intratumoral reactive oxygen species (ROS) are strongly enhanced, and adenosine triphosphate (ATP) is reduced under near-infrared (NIR) laser irradiation. Following a single-dose intravenous injection of LIP-IR-CAT, tumor hypoxia can be seriously attenuated, at the same time creating an opportunity to enhance the efficacy of PDT on the tumor. Our in vivo data show that the nanoreactor LIP-IR-CAT, in combination with just two short time NIR laser irradiation sessions, can effectively inhibit the growth of solid tumors without systemic toxicity.


Assuntos
Peróxido de Hidrogênio/administração & dosagem , Hipóxia/terapia , Neoplasias/terapia , Fármacos Fotossensibilizantes/administração & dosagem , Animais , Catalase/administração & dosagem , Catalase/química , Linhagem Celular Tumoral , Feminino , Humanos , Peróxido de Hidrogênio/química , Hipóxia/metabolismo , Indóis/administração & dosagem , Indóis/química , Raios Infravermelhos , Lipossomos/administração & dosagem , Lipossomos/química , Camundongos Endogâmicos BALB C , Nanopartículas/administração & dosagem , Nanopartículas/química , Neoplasias/metabolismo , Oxigênio/análise , Oxigênio/metabolismo , Fotoquimioterapia , Fármacos Fotossensibilizantes/química , Espécies Reativas de Oxigênio/metabolismo
12.
Mater Sci Eng C Mater Biol Appl ; 108: 110460, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31923975

RESUMO

Myelosuppression, gastrointestinal toxicity and hypersensitivities always accompany chemotherapy of osteosarcoma (OS). In addition, the intricate karyotype of OS, the lack of targeted antitumor drugs and the bone microenvironment that provides a protective alcove for tumor cells reduce the therapeutic efficacy of chemotherapy. Here, we developed a multifunctional bone cement loaded with Fe3O4 nanoparticles and the antitumor drug doxorubicin (DOX/Fe3O4@PMMA) for synergistic MH ablation and chemotherapy of OS. The localized intratumorally administered DOX/Fe3O4@PMMA can change from liquid into solid at the tumor site via a polyreaction. The designed multifunctional bone cement was constructed with Fe3O4 nanoparticles, PMMA, and an antitumor drug approved by the U.S. Food and Drug administration (FDA). The injectability, magnetic hyperthermia (MH) performance, controlled drug release profile, and synergistic therapeutic effect of DOX/Fe3O4@PMMA in vitro were investigated in detail. Furthermore, the designed DOX/Fe3O4@PMMA controlled the release of DOX, enhanced the apoptosis of OS tissue, and inhibited the proliferation of tumor cells, demonstrating synergistic MH ablation and chemotherapy of OS in vivo. The biosafety of DOX/Fe3O4@PMMA was also evaluated in detail. This strategy significantly reduced surgical time, avoided operative wounds and prevented patient pain, showing a great clinical translational potential for OS treatment.


Assuntos
Cimentos Ósseos , Neoplasias Ósseas/terapia , Hipertermia Induzida , Nanopartículas de Magnetita , Osteossarcoma/terapia , Animais , Cimentos Ósseos/química , Cimentos Ósseos/farmacologia , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Doxorrubicina/química , Doxorrubicina/farmacologia , Humanos , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Polimetil Metacrilato/química , Polimetil Metacrilato/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Adv Healthc Mater ; 8(13): e1900123, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30972958

RESUMO

Hypertrophic scarring (HS) remains a great challenge in wound dressing. Although various bionic extracellular matrix (ECM) biomaterials have been designed towards HS treatment, not all biomaterials can synergize biological functions and application functions in wound repair. Bionic scar-inhibiting scaffolds, loaded with biomolecules or drugs, become promising strategies for scarless skin regeneration. In this work, inspired by the physicochemical environment of ECM, a versatile fabrication of poly(γ-glutamic acid) based on electrospun photocrosslinkable hydrogel fibrous scaffolds incorporated with ginsenoside Rg3 (GS-Rg3) is developed for tissue repair and wound therapy. Decorated with adhesive peptide, bionic fibrous scaffolds can accelerate fibroblasts to sprout and grow, forming organized space-filling basement that gradually fills a depression before wound close up in the early stage. Additionally, by sustained release of GS-Rg3 in late stage, fibrous scaffolds promote scarless wound healing in vivo as evidenced by the promotion of cell communication and skin regeneration, as well as the subsequent decrease of angiogenesis and collagen accumulation. These ECM-inspired fibrous scaffolds, therefore, offer new perspectives on accelerated wound healing and tissue regeneration.


Assuntos
Cicatriz Hipertrófica/prevenção & controle , Ácido Poliglutâmico/análogos & derivados , Alicerces Teciduais/química , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Materiais Biocompatíveis/uso terapêutico , Linhagem Celular , Proliferação de Células , Cicatriz Hipertrófica/patologia , Liberação Controlada de Fármacos , Matriz Extracelular/química , Fibroblastos/citologia , Fibroblastos/metabolismo , Ginsenosídeos/química , Ginsenosídeos/farmacologia , Ginsenosídeos/uso terapêutico , Hidrogéis/química , Masculino , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Oligopeptídeos/uso terapêutico , Ácido Poliglutâmico/química , Ratos , Ratos Sprague-Dawley , Pele/patologia , Cicatrização/efeitos dos fármacos
14.
Theranostics ; 9(14): 4192-4207, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31281541

RESUMO

Background: Minimally invasive modalities are of great interest in the field of treating bone tumors. However, providing reliable mechanical support and fast killing of tumor cells to achieve rapid recovery of physical function is still challenging in clinical works. Methods: A material with two functions, mechanical support and magnetic thermal ablation, was developed from Fe3O4 nanoparticles (NPs) distributed in a polymethylmethacrylate (PMMA) bone cement. The mechanical properties and efficiency of magnetic field-induced thermal ablation were systematically and successfully evaluated in vitro and ex vivo. CT images and pathological examination were successfully applied to evaluate therapeutic efficacy with a rabbit bone tumor model. Biosafety evaluation was performed with a rabbit in vivo, and a cytotoxicity test was performed in vitro. Results: An NP content of 6% Fe3O4 (PMMA-6% Fe3O4, mFe: 0.01 g) gave the most suitable performance for in vivo study. At the 56-day follow-up after treatment, bone tumors were ablated without obvious side effects. The pathological examination and new bone formation in CT images clearly illustrate that the bone tumors were completely eliminated. Correspondingly, after treatment, the tendency of bone tumors toward metastasis significantly decreased. Moreover, with well-designed mechanical properties, PMMA-6%Fe3O4 implantation endowed tumor-bearing rabbit legs with excellent bio-mimic bone structure and internal support. Biosafety evaluation did not induce an increase or decrease in the immune response, and major functional parameters were all at normal levels. Conclusion: We have presented a novel, highly efficient and minimally invasive approach for complete bone tumor regression and bone defect repair by magnetic thermal ablation based on PMMA containing Fe3O4 NPs; this approach shows excellent heating ability for rabbit VX2 tibial plateau tumor ablation upon exposure to an alternating magnetic field (AMF) and provides mechanical support for bone repair. The new and powerful dual-function implant is a promising minimally invasive agent for the treatment of bone tumors and has good clinical translation potential.


Assuntos
Neoplasias Ósseas/terapia , Compostos Férricos/química , Polimetil Metacrilato/química , Animais , Hipertermia Induzida/métodos , Nanopartículas de Magnetita/química , Camundongos , Coelhos , Ratos
15.
Int J Nanomedicine ; 12: 3591-3603, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28533682

RESUMO

A polyethylene glycol-poly(ε-benzyloxycarbonyl-l-lysine) (PEG-SS-PLL) block copolymer based on a disulfide-linked, novel biodegradable catiomer bearing a PEG-sheddable shell was developed to avoid "PEG dilemma" in nanoparticle intracellular tracking of PEG-PLL where PEG was nondegradable. However, PEG-SS-PLL catiomers have not been used to deliver small interfering VEGF RNA (siVEGF) in antiangiogenesis gene therapy. In this study, we aimed to investigate whether this novel biodegradable catiomer can deliver siVEGF into cancer cells and at the same time have an antitumor effect in a xenograft mouse model. It was found that PEG-SS-PLL efficiently delivered siVEGF with negligible cytotoxicity, and significantly decreased the expression of VEGF at both the messenger-RNA and protein levels both in vitro and in vivo, and thus tumor growth was inhibited. Our findings demonstrated that PEG-SS-PLL/siVEGF could potentially be applied to antiangiogenesis gene therapy for hepatocellular carcinoma.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Terapia Genética/métodos , Polietilenoglicóis/química , Polilisina/análogos & derivados , RNA Interferente Pequeno/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/genética , Inibidores da Angiogênese/genética , Inibidores da Angiogênese/farmacologia , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Feminino , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Lisina/química , Camundongos Endogâmicos BALB C , Nanopartículas/administração & dosagem , Nanopartículas/química , Polilisina/química , Polímeros/química , Succinimidas/química , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Theranostics ; 7(18): 4410-4423, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29158836

RESUMO

Multifunctional nanoparticles have been reported for cancer detection and treatment currently. However, the accurate diagnosis and efficient treatment for tumors are still not satisfied. Here we report on the development of targeted phase change multimodal polymeric nanoparticles for the imaging and treatment of HER2-positive breast cancer. METHODS: We evaluated the multimodal imaging capabilities of the prepared nanoparticles in vitro using agar-based phantoms. The targeting performance and cytotoxicity of the nanoparticles were examined in cell culture using SKBR3 (over-expressing HER2) and MDA-MB-231 (HER2 negative) cells. We then tested the magnetic resonance (MR)/ photoacoustic (PA)/ ultrasound (US)/ near-infrared fluorescence (NIRF) multimodal imaging properties and photothermal effect of the nanoparticles in vivo using a SKBR3 breast xenograft model in nude mice. Tissue histopathology and immunofluorescence were also conducted. RESULTS: Both in vitro and in vivo systematical studies validated that the hybrid nanoparticles can be used as a superb MR/US/PA/NIRF contrast agent to simultaneously diagnose and guide tumor photothermal therapy (PTT). When irradiated by a near infrared laser, the liquid PFP vaporizes to a gas, rapidly expelling the contents and damaging surrounding tissues. The resulting micro-sized bubbles provide treatment validation through ultrasound imaging. Localization of DIR and SPIO in the tumor region facilitate photothermal therapy for targeted tumor destruction. The mice treated with HER2 targeted nanoparticles had a nearly complete response to treatment, while the controls showed continued tumor growth. CONCLUSION: This novel theranostic agent may provide better diagnostic imaging and therapeutic potential than current methods for treating HER2-positive breast cancer.


Assuntos
Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Neoplasias da Mama/terapia , Nanopartículas/administração & dosagem , Nanopartículas/química , Animais , Linhagem Celular Tumoral , Meios de Contraste/administração & dosagem , Feminino , Humanos , Lasers , Camundongos , Camundongos Nus , Imagem Multimodal/métodos , Fototerapia/métodos , Nanomedicina Teranóstica/métodos
17.
Biomaterials ; 128: 84-93, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28301803

RESUMO

The development of efficient strategies for the magnetic hyperthermia ablation of tumors remains challenging. To overcome the significant safety limitations, we developed a thermally contractible, injectable and biodegradable material for the minimally invasive and highly efficient magnetic hyperthermia ablation of tumors. This material was composed of hydroxypropyl methyl cellulose (HPMC), polyvinyl alcohol (PVA) and Fe3O4. The thermal contractibility of HPMC/Fe3O4 was designed to avoid damaging the surrounding normal tissue upon heating, which was confirmed by visual inspection, ultrasound imaging and computed tomography (CT). The efficient injectability of HPMC/Fe3O4 was proven using a very small needle. The biosafety of HPMC/Fe3O4 was evaluated by MTT and biochemical assays as well as flow cytometry (FCM). All the aforementioned data demonstrated the safety of HPMC/Fe3O4. The results of in vitro and ex vivo experiments showed that the temperature and necrotic volume of excised bovine liver were positively correlated with the HPMC/Fe3O4 weight, iron content and heating duration. The in vivo experimental results showed that the tumors could be completely ablated using 0.06 ml of HPMC/60%Fe3O4 after 180 s of induction heating. We believe that this novel, safe and biodegradable material will promote the rapid bench-to-bed translation of magnetic hyperthermia technology, and it is also expected to bring a new concept for the biomaterial research field.


Assuntos
Compostos Férricos/química , Hipertermia Induzida , Derivados da Hipromelose/química , Injeções , Fenômenos Magnéticos , Neoplasias/terapia , Temperatura , Animais , Bovinos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Compostos Férricos/toxicidade , Humanos , Derivados da Hipromelose/síntese química , Derivados da Hipromelose/toxicidade , Fígado/patologia , Camundongos Nus
18.
Methods Mol Biol ; 1364: 49-61, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26472441

RESUMO

Engineered PEG-cleavable catiomers based on poly-L-lysine have been developed as nonviral gene vectors, which have been found to be one of important methods to balance "PEG dilemma." In this protocol, we aim at the standardization of the method and procedure of PEG-cleavable catiomers. Major steps including ring-opening polymerization (ROP) of ε-benzyloxycarbonyl-L-lysine N-carboxyanhydride (zLL-NCA) monomers to yield PEG-cleavable polylysine, examination on bio-stability and bio-efficacy of its gene complexes are described.


Assuntos
Dissulfetos/química , Portadores de Fármacos/química , Polietilenoglicóis/química , Polilisina/química , RNA Interferente Pequeno/química , Transporte Biológico , Linhagem Celular , Portadores de Fármacos/metabolismo , Portadores de Fármacos/toxicidade , Estabilidade de Medicamentos , Humanos , Polilisina/metabolismo , Polilisina/toxicidade , Polimerização
19.
Adv Mater ; 28(10): 1963-9, 2016 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-26743228

RESUMO

A stability-difference-selective bond-breakage strategy for the fabrication of largepore-sized hollow mesoporous organosilica nanoparticles (HMONs) is successfully developed. Moreover, surfacefunctionalized HMONs are successfully constructed to simultaneously deliver P-gp modulator siRNA and anticancer drug doxorubicin to reverse the multidrug resistance of cancer cells.


Assuntos
Portadores de Fármacos/química , Técnicas de Transferência de Genes , Dióxido de Silício/química , Ligas/química , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Catálise , Humanos , Metais/química , Nanotubos/química , Neoplasias/tratamento farmacológico , Oxirredução , Óxidos/química , Espectroscopia Fotoeletrônica , Porosidade
20.
Acta Biomater ; 29: 298-306, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26432438

RESUMO

Magnetic hyperthermia ablation is a new and minimally invasive modality for localized tumor removal. However, an inadequate ablation dosage can leave a residual tumor or cause a variety of complications. In addition, commonly used magnetic nanoparticles can easily escape from the tumor tissue, which present potential safety problems. In this study, a smart phase transitional and injectable implant based on biocompatible poly lactic-co-glycolic acid (PLGA) implant incorporating magnetic material (Fe powder) and anti-cancer drug (doxorubicin (DOX)) was developed. The magnetic-induced hyperthermia and release efficiency of DOX were evaluated in vitro. Drug release can be controlled under external alternating current magnetic field (AMF). The results of the in vivo tumor therapeutic efficacy showed that when exposed to external AMF, this smart injectable DOX/PLGA-Fe implant could converse magnetic energy into heat and accelerate the release of DOX, which leads to increasing the temperature to achieve tumor coagulative necrosis and accelerating the release of DOX to enhance residual tumor apoptosis. Furthermore, there was no leakage of magnetic material, as demonstrated using real-time ultrasound (US) and computerized tomography (CT) imaging, realizing the guidance and monitoring of tumor therapy. In conclusion, this smart phase transitional and injectable implant DOX/PLGA-Fe has the ability to improve the efficiency of this newly developed minimally invasive magnetic ablation of tumor treatment technique, and will provide a new avenue of developing minimally invasive synergistic tumor therapy.


Assuntos
Doxorrubicina , Implantes de Medicamento , Hipertermia Induzida , Ferro , Ácido Láctico , Campos Magnéticos , Neoplasias Experimentais/terapia , Ácido Poliglicólico , Animais , Bovinos , Linhagem Celular Tumoral , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacologia , Implantes de Medicamento/química , Implantes de Medicamento/farmacocinética , Implantes de Medicamento/farmacologia , Humanos , Ferro/química , Ferro/farmacocinética , Ferro/farmacologia , Ácido Láctico/química , Ácido Láctico/farmacocinética , Ácido Láctico/farmacologia , Camundongos , Ácido Poliglicólico/química , Ácido Poliglicólico/farmacocinética , Ácido Poliglicólico/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA