Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Metabolism ; 146: 155656, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37419179

RESUMO

BACKGROUND AND AIMS: Hepatosteatosis is one of the early features of alcoholic liver disease (ALD) and pharmaceutical or genetic interfering of the development of hepatosteatosis will efficiently alleviate the progression of ALD. Currently, the role of histone methyltransferase Setdb1 in ALD is not yet well understood. METHOD: Lieber-De Carli diet mice model and NIAAA mice model were constructed to confirm the expression of Setdb1. The hepatocyte-specific Setdb1-knockout (Setdb1-HKO) mice was established to determine the effects of Setdb1 in vivo. Adenovirus-Setdb1 were produced to rescue the hepatic steatosis in both Setdb1-HKO and Lieber-De Carli mice. The enrichment of H3k9me3 in the upstream sequence of Plin2 and the chaperone-mediated autophagy (CMA) of Plin2 were identified by ChIP and co-IP. Dual-luciferase reporter assay was used to detect the interaction of Setdb1 3'UTR and miR216b-5p in AML12 or HEK 293 T cells. RESULTS: We found that Setdb1 was downregulated in the liver of alcohol-fed mice. Setdb1 knockdown promoted lipid accumulation in AML12 hepatocytes. Meanwhile, hepatocyte-specific Setdb1-knockout (Setdb1-HKO) mice exhibited significant lipid accumulation in the liver. Overexpression of Setdb1 was performed with an adenoviral vector through tail vein injection, which ameliorated hepatosteatosis in both Setdb1-HKO and alcoholic diet-fed mice. Mechanistically, downregulated Setdb1 promoted the mRNA expression of Plin2 by desuppressing H3K9me3-mediated chromatin silencing in its upstream sequence. Pin2 acts as a critical membrane surface-associated protein to maintain lipid droplet stability and inhibit lipase degradation. The downregulation of Setdb1 also maintained the stability of Plin2 protein through inhibiting Plin2-recruited chaperone-mediated autophagy (CMA). To explore the reasons for Setdb1 suppression in ALD, we found that upregulated miR-216b-5p bound to the 3'UTR of Setdb1 mRNA, disturbed its mRNA stability, and eventually aggravated hepatic steatosis. CONCLUSIONS: Setdb1 suppression plays an important role in the progression of alcoholic hepatosteatosis via elevating the expression of Plin2 mRNA and maintaining the stability of Plin2 protein. Targeting hepatic Setdb1 might be a promising diagnostic or therapeutic strategy for ALD.


Assuntos
Fígado Gorduroso , Hepatopatias Alcoólicas , Animais , Humanos , Camundongos , Regiões 3' não Traduzidas , Fígado Gorduroso/metabolismo , Células HEK293 , Lipídeos , Fígado/metabolismo , Hepatopatias Alcoólicas/metabolismo , Perilipina-2/genética , Perilipina-2/metabolismo
2.
Biomaterials ; 296: 122090, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36940634

RESUMO

Therapeutic gas molecules have high tissue penetrability, but their sustainable supply and controlled release in deep tumor is a huge challenge. In this work, a concept of sonocatalytic full water splitting for hydrogen/oxygen immunotherapy of deep tumor is proposed, and a new kind of ZnS nanoparticles with a mesocrystalline structure (mZnS) is developed to achieve highly efficient sonocatalytic full water splitting for sustainable supply of H2 and O2 in tumor, achieving a high efficacy of deep tumor therapy. Mechanistically, locally generated hydrogen and oxygen molecules exhibit a tumoricidal effect as well as the co-immunoactivation of deep tumors through inducing the M2-to-M1 repolarization of intratumoral macrophages and the tumor hypoxia relief-mediated activation of CD8+ T cells, respectively. The proposed sonocatalytic immunoactivation strategy will open a new window to realize safe and efficient treatment of deep tumors.


Assuntos
Nanopartículas , Neoplasias , Humanos , Água , Linfócitos T CD8-Positivos , Nanopartículas/química , Neoplasias/terapia , Oxigênio/uso terapêutico , Hidrogênio/uso terapêutico , Linhagem Celular Tumoral , Microambiente Tumoral
3.
Bioact Mater ; 14: 31-41, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35310347

RESUMO

Engineering biomaterials to meet specific biomedical applications raises high requirements of mechanical performances, and simultaneous strengthening and toughening of polymer are frequently necessary but very challenging in many cases. In this work, we propose a new concept of nanoconcrete welding polymer chains, where mesoporous CaCO3 (mCaCO3) nanoconcretes which are composed of amorphous and nanocrystalline phases are developed to powerfully weld polymer chains through siphoning-induced occlusion, hydration-driven crystallization and dehydration-driven compression of nanoconcretes. The mCaCO3 nanoconcrete welding technology is verified to be able to remarkably augment strength, toughness and anti-fatigue performances of a model polymer poly(3-hydroxybutyrate-co-3-hydroxyvalerate)-based porous membrane. Mechanistically, we have revealed polymer-occluded nanocrystal structure and welding-derived microstress which is much stronger than interfacial Van der Waals force, thus efficiently preventing the generation of microcracks and repairing initial microcracks by microcracks-induced hydration, crystallization and polymer welding of mCaCO3 nanoconcretes. Constructed porous membrane is used as wound dressing, exhibiting a special nanoplates-constructed surface topography as well as a porous structure with plentiful oriented, aligned and opened pore channels, improved hydrophilicity, water vapor permeability, anti-bacterial and cell adherence, in support of wound healing and skin structural/functional repairing. The proposed nanoconcrete-welding-polymer strategy breaks a new pathway for improving the mechanical performances of polymers.

4.
Bioact Mater ; 12: 303-313, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35128178

RESUMO

Tumor-targeted delivery of nanomedicine is of great importance to improve therapeutic efficacy of cancer and minimize systemic side effects. Unfortunately, nowadays the targeting efficiency of nanomedicine toward tumor is still quite limited and far from clinical requirements. In this work, we develop an innovative peptide-based nanoparticle to realize light-triggered nitric oxide (NO) release and structural transformation for enhanced intratumoral retention and simultaneously sensitizing photodynamic therapy (PDT). The designed nanoparticle is self-assembled from a chimeric peptide monomer, TPP-RRRKLVFFK-Ce6, which contains a photosensitive moiety (chlorin e6, Ce6), a ß-sheet-forming peptide domain (Lys-Leu-Val-Phe-Phe, KLVFF), an oligoarginine domain (RRR) as NO donor and a triphenylphosphonium (TPP) moiety for targeting mitochondria. When irradiated by light, the constructed nanoparticles undergo rapid structural transformation from nanosphere to nanorod, enabling to achieve a significantly higher intratumoral accumulation by 3.26 times compared to that without light irradiation. More importantly, the conversion of generated NO and reactive oxygen species (ROS) in a light-responsive way to peroxynitrite anions (ONOO-) with higher cytotoxicity enables NO to sensitize PDT in cancer treatment. Both in vitro and in vivo studies demonstrate that NO sensitized PDT based on the well-designed transformable nanoparticles enables to eradicate tumors efficiently. The light-triggered transformable nanoplatform developed in this work provides a new strategy for enhanced intratumoral retention and improved therapeutic outcome.

5.
Adv Sci (Weinh) ; 9(10): e2101965, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35098699

RESUMO

The development of stimuli-responsively degradable porous carriers for both controlled drug release and high biosafety is vitally important to their clinical translation, but still challenging at present. A new type of porphyrin-iron metal organic framework (Fe-MOF) nanocrystals is engineered here as acid-degradable drug carrier and hydrogen donor by the coordination between porphyrin and zero-valence Fe atom. Fe-MOF nanocrystals exhibit excellent acid-responsive degradation for H2 generation and simultaneous release of the loaded drug for combined hydrogen-chemotherapy of cancer multidrug resistance (MDR) and metastasis and for local hydrogen eradication of the off-target induced toxic side effects of the drug to normal cells/tissues. Mechanistically, released H2 assists chemotherapeutic drug to efficiently inhibit cancer metastasis by immunoactivating intratumoral M1-phenotype macrophages and consequently downregulating the expression of metastasis-related matrix metalloproteinase-2 (MMP-2) and can also downregulate the expressions of both P-glycoprotein (P-gp) protein and adenosine triphosphate (ATP) in MDR cancer cells to sensitize chemotherapeutic drug for enhanced damage to mitochondria and DNA. High anti-MDR/antimetastasis efficacies and high biocompatibility endow Fe-MOF nanocrystals and the Fe-MOF-based nanomedicine with high potential for clinical translation.


Assuntos
Estruturas Metalorgânicas , Neoplasias , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Resistência a Múltiplos Medicamentos , Hidrogênio/farmacologia , Metaloproteinase 2 da Matriz/farmacologia , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/farmacologia , Neoplasias/tratamento farmacológico
6.
Adv Mater ; 33(39): e2101455, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34369623

RESUMO

The efficient utilization of near-infrared (NIR) light for photocatalytic hydrogen generation is vitally important to both solar hydrogen energy and hydrogen medicine, but remains a challenge at present, owing to the strict requirement of the semiconductor for high NIR responsiveness, narrow bandgap, and suitable redox potentials. Here, an NIR-active carbon/potassium-doped red polymeric carbon nitride (RPCN) is achieved for by using a similar-structure dopant as the melamine (C3 H6 N6 ) precursor with the solid KCl. The homogeneous and high incorporation of carbon and potassium remarkably narrows the bandgap of carbon nitride (1.7 eV) and endows RPCN with a high NIR-photocatalytic activity for H2 evolution from water at the rate of 140 µmol h-1 g-1 under NIR irradiation (700 nm ≤ λ ≤ 780 nm), and the apparent quantum efficiency is high as 0.84% at 700 ± 10 nm (and 13% at 500 ± 10 nm). A proof-of-concept experiment on a tumor-bearing mouse model verifies RPCN as being capable of intratumoral NIR-photocatalytic hydrogen generation and simultaneous glutathione deprivation for safe and high-efficacy drug-free cancer therapy. The results shed light on designing efficient photocatalysts to capture the full spectrum of solar energy, and also pioneer a new pathway to develop NIR photocatalysts for hydrogen therapy of major diseases.

7.
ACS Appl Mater Interfaces ; 13(24): 27963-27971, 2021 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-34110773

RESUMO

1T-phase transition-metal dichalcogenides (TMDs) nanomaterials are one type of emerging and promising near-infrared II (NIR-II) photothermal agents (PTAs) derived from their distinct metallic electronic structure, but it is still challenging to synthesize these nanomaterials. Herein, PdTe2 nanoparticles (PTNs) with a 1T crystal symmetry and around 50 nm in size are prepared by an electrochemical exfoliation method, and the corresponding photothermal performances irradiated under a NIR-II laser have been explored. The encapsulation of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-poly(ethylene glycol) (DSPE-PEG) endows PTNs with water solubility, enhanced photothermal stability, and high biocompatibility. Notably, PTN/DSPE-PEG displays a potent absorbance through the NIR-II zone and considerable photothermal conversion efficiency, which is up to 68% when irradiated with a 1060 nm laser. With these unique photothermal properties, excellent in vitro and in vivo tumor inhibition effects of PTN/DSPE-PEG have been achieved under the irradiation of a NIR-II (1060 nm) laser without visible toxicity to normal tissues, suggesting that it is an efficient NIR-II photothermal nanoagent.


Assuntos
Antineoplásicos/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/química , Antineoplásicos/efeitos da radiação , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Feminino , Interações Hidrofóbicas e Hidrofílicas , Raios Infravermelhos , Nanopartículas Metálicas/química , Nanopartículas Metálicas/efeitos da radiação , Nanopartículas Metálicas/toxicidade , Camundongos Endogâmicos BALB C , Paládio/química , Paládio/efeitos da radiação , Paládio/uso terapêutico , Paládio/toxicidade , Fosfatidiletanolaminas/química , Terapia Fototérmica , Polietilenoglicóis/química , Telúrio/química , Telúrio/efeitos da radiação , Telúrio/uso terapêutico , Telúrio/toxicidade
8.
Nat Commun ; 12(1): 1345, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33649319

RESUMO

Drug therapy unavoidably brings toxic side effects and drug content-limited therapeutic efficacy although many nanocarriers have been developed to improve them to a certain extent. In this work, a concept of drug-free therapeutics is proposed and defined as a therapeutic methodology without the use of traditional toxic drugs, without the consumption of therapeutic agents during treatment but with the inexhaustible therapeutic capability to maximize the benefit of treatment, and a Z-scheme SnS1.68-WO2.41 nanocatalyst is developed to achieve near infrared (NIR)-photocatalytic generation of oxidative holes and hydrogen molecules for realizing combined hole/hydrogen therapy by the drug-free therapeutic strategy. Without the need of any drug and other therapeutic agent assistance, the nanocatalyst oxidizes/consumes intratumoral over-expressed glutathione (GSH) by holes and simultaneously generates hydrogen molecules in a lasting and controllable way under NIR irradiation. Mechanistically, generated hydrogen molecules and GSH consumption inhibit cancer cell energy and destroy intratumoral redox balance, respectively, to synergistically damage DNA and induce tumor cell apoptosis. High efficacy and biosafety of combined hole/hydrogen therapy of tumors are achieved by the nanocatalyst. The proposed catalysis-based drug-free therapeutic strategy breaks a pathway to realize high efficacy and low toxicity of cancer treatment.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Fototerapia , Animais , Catálise/efeitos da radiação , Linhagem Celular Tumoral , Glutationa/química , Humanos , Hidrogênio/química , Raios Infravermelhos , Antígeno Ki-67/metabolismo , Camundongos , Nanopartículas/ultraestrutura , Tamanho da Partícula , Análise Espectral , Carga Tumoral , Microambiente Tumoral
9.
Sci Adv ; 6(20): eaba1362, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32440551

RESUMO

CO gas molecule not only could selectively kill cancer cells but also exhibits limited anticancer efficacy because of the lack of active tumor-targeted accumulation capability. In this work, a multistage assembly/disassembly strategy is developed to construct a new intelligent nanomedicine by encapsulating a mitochondria-targeted and intramitochondrial microenvironment-responsive prodrug (FeCO-TPP) within mesoporous silica nanoparticle that is further coated with hyaluronic acid by step-by-step electrostatic assembly, realizing tumor tissue-cell-mitochondria-targeted multistage delivery and controlled release of CO in a step-by-step disassembly way. Multistage targeted delivery and controlled release of CO involve (i) the passive tumor tissue-targeted nanomedicine delivery, (ii) the active tumor cell-targeted nanomedicine delivery, (iii) the acid-responsive prodrug release, (iv) the mitochondria-targeted prodrug delivery, and (v) the ROS-responsive CO release. The developed nanomedicine has effectively augmented the efficacy and safety of CO therapy of cancer both in vitro and in vivo. The proposed multistage assembly/disassembly strategy opens a new window for targeted CO therapy.


Assuntos
Nanopartículas , Neoplasias , Pró-Fármacos , Linhagem Celular Tumoral , Preparações de Ação Retardada , Sistemas de Liberação de Medicamentos , Humanos , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Dióxido de Silício , Microambiente Tumoral
10.
Theranostics ; 10(4): 1861-1872, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32042341

RESUMO

Background: Rapid advance in biomedicine has recently vitalized the development of multifunctional two-dimensional (2D) nanomaterials for cancer theranostics. However, it is still challenging to develop new strategy to produce new types of 2D nanomaterials with flexible structure and function for enhanced disease theranostics. Method: We explore the monolayer Bi-anchored manganese boride nanosheets (MBBN) as a new type of MBene (metal boride), and discover their unique near infrared (NIR)-photothermal and photoacoustic effects, X-ray absorption and MRI imaging properties, and develop them as a new nanotheranostic agent for multimodal imaging-guided photothermal therapy of cancer. A microwave-assisted chemical etching route was utilized to exfoliate the manganese boride bulk into the nanosheets-constructed flower-like manganese boride nanoparticle (MBN), and a coordination-induced exfoliation strategy was further developed to separate the MBN into the dispersive monolayer MBBN by the coordination between Bi and B on the surface, and the B-OH group on the surface of MBBN enabled facile surface modification with hyaluronic acid (HA) by the borate esterification reaction in favor of enhanced monodispersion and active tumor targeting. Result: The constructed MBBN displays superior NIR-photothermal conversion efficiency (η=59.4%) as well as high photothermal stability, and possesses versatile imaging functionality including photoacoustic, photothermal, CT and T1 -wighted MRI imagings. In vitro and in vivo evaluations indicate that MBBN had high photothermal ablation and multimodal imaging performances, realizing high efficacy of imaging-guided cancer therapy. Conclusion: We have proposed new MBene concept and exfloliation strategy to impart the integration of structural modification and functional enhancement for cancer theranostics, which would open an avenue to facile fabrication and extended application of multifunctional 2D nanomaterials.


Assuntos
Nanopartículas Metálicas/química , Imagem Multimodal/métodos , Neoplasias/diagnóstico por imagem , Terapia Fototérmica/métodos , Nanomedicina Teranóstica/métodos , Animais , Feminino , Humanos , Imageamento por Ressonância Magnética/métodos , Compostos de Manganês/química , Compostos de Manganês/metabolismo , Compostos de Manganês/farmacologia , Camundongos , Camundongos Nus , Modelos Animais , Nanopartículas/química , Nanoestruturas/química , Neoplasias/terapia , Técnicas Fotoacústicas/métodos
11.
J Nanobiotechnology ; 17(1): 75, 2019 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-31196217

RESUMO

BACKGROUND: To improve the outcome of cancer treatment, the combination of multiple therapy models has proved to be effective and promising. Gas therapy (GT) and chemodynamic therapy (CDT), mainly targeting the mitochondrion and nucleus, respectively, are two emerging strategy for anti-cancer. The development of novel nanomedicine for integrating these new therapy models is greatly significant and highly desired. METHODS: A new nanomedicine is programmed by successive encapsulation of MnO2 nanoparticles and iron carbonyl (FeCO) into mesoporous silica nanoparticle. By decoding the nanomedicine, acidity in the lysosome drives MnO2 to generate ROS, ·OH among which further triggers the decomposition of FeCO into CO, realizing the effective combination of chemodynamic therapy with gas therapy for the first time. RESULTS: Acidity in the TEM drives MnO2 to generate ROS, ∙OH among which further triggers the decomposition of FeCO into CO, realizing the effective combination of CDT and CDGT. The co-released ROS and CO do damage to DNA and mitochondria of various cancer cells, respectively. The mitochondrial damage can effectively cut off the ATP source required for DNA repair, causing a synergetic anti-cancer effect in vitro and in vivo. CONCLUSIONS: The combination of CDT and CDGT causing a synergetic anti-cancer effect in vitro and in vivo. The proposed therapy concept and nanomedicine designing strategy might open a new window for engineering high-performance anti-cancer nanomedicine.


Assuntos
Monóxido de Carbono/química , Compostos Carbonílicos de Ferro/química , Compostos de Manganês/química , Nanopartículas/química , Óxidos/química , Espécies Reativas de Oxigênio/química , Dióxido de Silício/química , Animais , Monóxido de Carbono/administração & dosagem , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Camundongos Endogâmicos BALB C , Nanomedicina , Oxirredução , Porosidade , Espécies Reativas de Oxigênio/administração & dosagem
12.
Adv Healthc Mater ; 8(13): e1900157, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30968583

RESUMO

The hydrogen molecule is recognized as a high potential to attenuate toxic side effects of chemotherapy and also enhance chemotherapeutic efficacy, and the development of a novel hydrogen-generating prodrug for facile, safe, and efficient hydrogen delivery is vitally important for combined hydrogenochemotherapy but is still challenging. Here, targeting gastric cancer, a 2D magnesium boride nanosheet (MBN) is synthesized as a new type of acid-responsive hydrogen-releasing prodrug by an ultrasound-assisted chemical etching route, which is used to realize hydrogenochemotherapy by combination of facile oral administration of polyvinylpyrrolidone (PVP)-encapsulating MBN (MBN@PVP) pills with routine intravenous injection of doxorubicin (DOX). The MBN@PVP pill has high stability in normal tissues/blood environments as well as high gastric acid-responsiveness with sustained release behavior, which matches well with its metabolism rate in the stomach in great favor of continuous and long-term hydrogen administration. Hydrogenochemotherapy with DOX+MBN@PVP has remarkably prolonged the survival time of gastric tumor-bearing mice by reducing the toxic side effects of chemotherapy. The mechanism for therapeutic synergy and side effect attenuation of hydrogenochemotherapy is discovered to be derived from the selectivity of hydrogen molecules in inhibiting aerobic respiration of gastric cells but activating aerobic respiration of normal cells including marrow mesenchymal stem cells and cardiac, hepatic, and splenic cells.


Assuntos
Compostos de Boro/química , Hidrogênio/metabolismo , Compostos de Magnésio/química , Nanoestruturas/química , Pró-Fármacos/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Administração Oral , Animais , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular , Doxorrubicina/química , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Nus , Povidona/química , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Neoplasias Gástricas/mortalidade , Neoplasias Gástricas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA