Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Nanotechnol ; 2024 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-39327512

RESUMO

The success of personalized cancer immunotherapy depends on the initial tumour antigenic presentation to dendritic cells and macrophages. Tumour-derived extracellular vesicles (TEVs) contain abundant tumour antigenic molecules. The presence of anti-phagocytotic signals such as cluster of differentiation 47 (CD47) on the surface of the TEVs, however, leads to evasion of the same dendritic cells and macrophages. Here we show that iron oxide hydroxide nanocomposites can successfully mask TEV surfaces and unblock phagocytosis without affecting extracellular vesicles' elicited immune goals. After internalization, the mask disintegrates in the lysosome, releasing the tumour antigenic cargo. This triggers antigen presentation and promotes dendritic cell activation and maturation and macrophage reprogramming in animal models, leading to a drastic reduction of tumour volume and metastasis, and in human malignant pleural effusion clinical samples. This straightforward masking strategy eliminates the ubiquitous anti-phagocytosis block found in clinical samples and can be applied universally across all patient-specific TEVs as tumour antigenic agents for enhanced immunotherapy.

2.
Nanoscale Horiz ; 9(7): 1190-1199, 2024 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-38757185

RESUMO

Antibacterial nanoagents have been increasingly developed due to their favorable biocompatibility, cost-effective raw materials, and alternative chemical or optical properties. Nevertheless, there is still a pressing need for antibacterial nanoagents that exhibit outstanding bacteria-binding capabilities and high antibacterial efficiency. In this study, we constructed a multifunctional cascade bioreactor (GCDCO) as a novel antibacterial agent. This involved incorporating carbon dots (CDs), cobalt sulfide quantum dots (CoSx QDs), and glucose oxidase (GOx) to enhance bacterial inhibition under sunlight irradiation. The GCDCO demonstrated highly efficient antibacterial capabilities attributed to its favorable photothermal properties, photodynamic activity, as well as the synergistic effects of hyperthermia, glucose-augmented chemodynamic action, and additional photodynamic activity. Within this cascade bioreactor, CDs played the role of a photosensitizer for photodynamic therapy (PDT), capable of generating ˙O2- even under solar light irradiation. The CoSx QDs not only functioned as a catalytic component to decompose hydrogen peroxide (H2O2) and generate hydroxyl radicals (˙OH), but they also served as heat generators to enhance the Fenton-like catalysis process. Furthermore, GOx was incorporated into this cascade bioreactor to internally supply H2O2 by consuming glucose for a Fenton-like reaction. As a result, GCDCO could generate a substantial amount of reactive oxygen species (ROS), leading to a significant synergistic effect that greatly induced bacterial death. Furthermore, the in vitro antibacterial experiment revealed that GCDCO displayed notably enhanced antibacterial activity against E. coli (99+ %) when combined with glucose under simulated sunlight, surpassing the efficacy of the individual components. This underscores its remarkable efficiency in combating bacterial growth. Taken together, our GCDCO demonstrates significant potential for use in the routine treatment of skin infections among diabetic patients.


Assuntos
Antibacterianos , Glucose Oxidase , Fotoquimioterapia , Pontos Quânticos , Pontos Quânticos/química , Pontos Quânticos/efeitos da radiação , Glucose Oxidase/química , Fotoquimioterapia/métodos , Antibacterianos/farmacologia , Antibacterianos/química , Escherichia coli/efeitos dos fármacos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/química , Cobalto/química , Cobalto/farmacologia , Luz , Carbono/química , Carbono/farmacologia , Humanos , Peróxido de Hidrogênio/farmacologia , Reatores Biológicos , Espécies Reativas de Oxigênio/metabolismo
3.
Nat Nanotechnol ; 19(1): 95-105, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37709950

RESUMO

An effective nanotherapeutic transport from the vasculature to the tumour is crucial for cancer treatment with minimal side effects. Here we demonstrate that, in addition to the endothelial barrier, the tumour vascular basement membrane surrounding the endothelium acts as a formidable mechanical barrier that entraps nanoparticles (NPs) in the subendothelial void, forming perivascular NP pools. Breaking through this basement membrane barrier substantially increases NP extravasation. Using inflammation triggered by local hyperthermia, we develop a cooperative immunodriven strategy to overcome the basement membrane barrier that leads to robust tumour killing. Hyperthermia-triggered accumulation and inflammation of platelets attract neutrophils to the NP pools. The subsequent movement of neutrophils through the basement membrane can release the NPs entrapped in the subendothelial void, resulting in increased NP penetration into deeper tumours. We show the necessity of considering the tumour vascular basement membrane barrier when delivering nanotherapeutics. Understanding this barrier will contribute to developing more effective antitumour therapies.


Assuntos
Neoplasias , Humanos , Membrana Basal/patologia , Neoplasias/patologia , Neutrófilos , Inflamação/patologia
4.
Nat Commun ; 14(1): 4269, 2023 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-37460554

RESUMO

Delivering cancer therapeutics to tumors necessitates their escape from the surrounding blood vessels. Tumor vasculatures are not always sufficiently leaky. Herein, we engineer therapeutically competent leakage of therapeutics from tumor vasculature with gold nanoparticles capable of inducing endothelial leakiness (NanoEL). These NanoEL gold nanoparticles activated the loss of endothelial adherens junctions without any perceivable toxicity to the endothelial cells. Microscopically, through real time live animal intravital imaging, we show that NanoEL particles induced leakiness in the tumor vessels walls and improved infiltration into the interstitial space within the tumor. In both primary tumor and secondary micrometastases animal models, we show that pretreatment of tumor vasculature with NanoEL particles before therapeutics administration could completely regress the cancer. Engineering tumoral vasculature leakiness represents a new paradigm in our approach towards increasing tumoral accessibility of anti-cancer therapeutics instead of further increasing their anti-cancer lethality.


Assuntos
Nanopartículas Metálicas , Nanopartículas , Neoplasias de Tecido Vascular , Neoplasias , Animais , Células Endoteliais/patologia , Ouro , Nanopartículas Metálicas/uso terapêutico , Endotélio/patologia , Neoplasias/patologia
5.
Nanoscale Horiz ; 8(2): 270-278, 2023 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-36598052

RESUMO

Vascular endothelium dysfunction plays an important role in oncological and pulmonary diseases. Endothelial barrier dysfunction is the initial step of pulmonary vascular remodeling (PVR) and pulmonary arterial hypertension. Upregulation of a pro-autophagy protein Atg101 in the endothelial cells triggered a cascade of intracellular events that leads to endothelial dysfunction through apoptosis. Herein, we proposed a strategy that used endothelial targeting DNA nanostructures to deliver Atg101 siRNA (siAtg101) as a safe, biocompatible "band-aid" to restore pulmonary arterial endothelial barrier integrity within the intricate milieu of pulmonary cells and the pulmonary vasculature. The siAtg101 and aptamer conjugated DNA nanostructures were found to attenuate hypoxia-induced pulmonary endothelial leakiness with surprisingly high selectivity and efficacy. Further in vivo study revealed that functionalized DNA nanostructures likewise attenuated the vascular remodeling in a monocrotaline-induced PVR mouse model. Mechanistically, functionalized DNA nanostructures suppressed PVR by knocking down Atg101, which in turn, downregulated Beclin-1 and subsequently upregulated VE-cadherin to restore endothelial cells' adherin junctions. This work opened a new window for future nanomaterial design that directly addresses the interfacial endothelial cell layer that often stands between the blood and many diseased sites of nanotherapeutic interest.


Assuntos
Hipertensão Pulmonar , Nanoestruturas , Hipertensão Arterial Pulmonar , Camundongos , Animais , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/metabolismo , Células Endoteliais , Remodelação Vascular , Hipertensão Pulmonar Primária Familiar , DNA/genética , DNA/uso terapêutico
6.
Nat Commun ; 13(1): 7289, 2022 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-36435865

RESUMO

The transfer of the concept of chirality from molecules to synthesized nanomaterials has attracted attention amongst multidisciplinary teams. Here we demonstrate heterogeneous nucleation and anisotropic accumulation of Au nanoparticles on multilayer MoS2 planes to form chiroptically functional nanomaterials. Thiol amino acids with chiral conformations modulate asymmetric growth of gold nanoarchitectures on seeds of highly faceted Au/MoS2 heterostructures. Consequently, dendritic plasmonic nanocrystals with partial chiral morphologies are synthesized. The chirality of dendritic nanocrystals inherited from cysteine molecules refers to the structural characteristics and includes specific recognition of enantiomeric molecules. With integration of the intrinsic photothermal properties and inherited enantioselective characteristics, dendritic Au/MoS2 heterostructures exhibit chirality-dependent release of antimicrobial drugs from hydrogel substrates when activated by exogenous infrared irradiation. A three-in-one strategy involving synthesis of chiral dendritic heterostructures, enantioselective recognition, and controlled drug release system is presented, which improves nanomaterial synthetic technology and enhances our understanding of crucial chirality information.


Assuntos
Anti-Infecciosos , Nanopartículas Metálicas , Ouro/química , Estereoisomerismo , Nanopartículas Metálicas/química , Molibdênio , Anti-Infecciosos/farmacologia
7.
Biomaterials ; 287: 121640, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35772348

RESUMO

All intravenous delivered nanomedicine needs to escape from the blood vessel to exert their therapeutic efficacy at their designated site of action. Failure to do so increases the possibility of detrimental side effects and negates their therapeutic intent. Many powerful anticancer nanomedicine strategies rely solely on the tumor derived enhanced permeability and retention (EPR) effect for the only mode of escaping from the tumor vasculature. However, not all tumors have the EPR effect nor can the EPR effect be induced or controlled for its location and timeliness. In recent years, there have been exciting developments along the lines of inducing endothelial leakiness at the tumor to decrease the dependence of EPR. Physical disruption of the endothelial-endothelial cell junctions with coordinated biological intrinsic pathways have been proposed that includes various modalities like ultrasound, radiotherapy, heat and even nanoparticles, appear to show good progress towards the goal of inducing endothelial leakiness. This review explains the intricate and complex biological background behind the endothelial cells with linkages on how updated reported nanomedicine strategies managed to induce endothelial leakiness. This review will also end off with fresh insights on where the future of inducible endothelial leakiness holds.

8.
ACS Appl Mater Interfaces ; 14(22): 25183-25196, 2022 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-35638599

RESUMO

Chemodynamic therapy (CDT), as a powerful tumor therapeutic approach with low side effects and selective therapeutic efficiency, has gained much attention. However, the low intracellular content of H2O2 and the cellular bottleneck of low intracellular oxidative reaction rates at tumor sites have limited the antitumor efficacy of CDT. Herein, a series of sulfur-deficient engineered biodegradable cobalt sulfide quantum dots (CoSx QDs) were constructed for improved synergistic photothermal- and hyperthermal-enhanced CDT of tumors through regulating the photothermal conversion efficiency (PCE) and Fenton-like activity. Through defect engineering, we modulated the PCE and promoted the Fenton catalytic capability of CoSx QDs. With increasing defect sites, the Fenton-like activity improved to generate more toxic •OH, while the photothermal effect declined slightly. In light of above unique superiorities, the best synergistic effects of CoSx QDs were obtained through comparing their PCE and catalytic activity by regulating the sulfur defect fraction degree in these QDs during the synthetic process. In addition, the ultrasmall size and biodegradation endowed QDs with the ability to be rapidly decomposed to ions that were easily excreted after therapy, thus reducing biogenic accumulation in the body with lowered systemic side effects. The in vitro/vivo results demonstrated that the photothermal- and hyperthermal-enhanced chemodynamic effect of CoSx QDs can enable remarkable anticancer properties with favorable biocompatibility. In this study, the defect-driven mechanism for the photothermal-enhanced Fenton-like reaction provides a flexible strategy to deal with different treatment environments, holding great promise in developing a multifunctional platform for cancer treatment in the future.


Assuntos
Nanopartículas , Neoplasias , Pontos Quânticos , Linhagem Celular Tumoral , Cobalto , Humanos , Peróxido de Hidrogênio/metabolismo , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Enxofre
9.
ACS Appl Mater Interfaces ; 13(50): 60351-60361, 2021 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-34874695

RESUMO

Transition-metal dichalcogenide (TMD)-based nanomaterials have been extensively explored for the photonic therapy. To the best of our knowledge, near-infrared (NIR) light is a requirement for the photothermal therapy (PTT) to achieve the feature of deep-tissue penetration, whereas no obvious absorption peaks existing in the NIR region for existing TMD nanomaterials limit their therapeutic efficacy. As one category of TMD nanomaterials, ruthenium sulfide-based nanomaterials have been less exploited in biomedical applications including tumor therapy so far. Here, we develop a facile biomineralization-assisted bottom-up strategy to synthesize oxygenic hybrid ruthenium sulfide nanoclusters (RuSx NCs) by regulating the oxygen amounts and sulfur defects for the optimized PTT. By regulating the increasing initial molar ratios of Ru to S, RuSx NCs with small sizes were endowed with increasing oxygen contents and sulfur defects, leading to the photothermal conversion efficiency (PCE) increasing from 32.8 to 41.9%, which were higher than that of most small-sized inorganic photothermal nanoagents. In contrast to commercial indocyanine green, these RuSx NCs exhibited higher photostability under NIR laser irradiation. The high PCE and superior photostability allowed RuSx NCs to effectively and completely ablate cancer cells. Thus, the proposed defect engineering strategy endows RuSx NCs with an excellent photothermal effect for the PTT of tumors of living mice, which also proves the potential of further exploring the properties of RuSx NCs for future biomedical applications.


Assuntos
Antineoplásicos/farmacologia , Materiais Biocompatíveis/farmacologia , Nanoestruturas/química , Oxigênio/farmacologia , Terapia Fototérmica , Rutênio/farmacologia , Sulfetos/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Materiais Biocompatíveis/síntese química , Materiais Biocompatíveis/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Raios Infravermelhos , Teste de Materiais , Oxigênio/química , Tamanho da Partícula , Rutênio/química , Sulfetos/química
10.
Adv Sci (Weinh) ; 8(21): e2102519, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34495564

RESUMO

Nanomaterial-induced endothelial leakiness (NanoEL) is an interfacial phenomenon denoting the paracellular transport of nanoparticles that is pertinent to nanotoxicology, nanomedicine and biomedical engineering. While the NanoEL phenomenon is complementary to the enhanced permeability and retention effect in terms of their common applicability to delineating the permeability and behavior of nanoparticles in tumoral environments, these two effects significantly differ in scope, origin, and manifestation. In the current study, the descriptors are fully examined of the NanoEL phenomenon elicited by generic citrate-coated gold nanoparticles (AuNPs) of changing size and concentration, from microscopic gap formation and actin reorganization down to molecular signaling pathways and nanoscale interactions of AuNPs with VE-cadherin and its intra/extracellular cofactors. Employing synergistic in silico methodologies, for the first time the molecular and statistical mechanics of cadherin pair disruption, especially in response to AuNPs of the smallest size and highest concentration are revealed. This study marks a major advancement toward establishing a comprehensive NanoEL framework for complementing the understanding of the transcytotic pathway and for guiding the design and application of future nanomedicines harnessing the myriad functions of the mammalian vasculature.


Assuntos
Ouro/química , Nanopartículas Metálicas/química , Animais , Antígenos CD/química , Antígenos CD/metabolismo , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/metabolismo , Caderinas/química , Caderinas/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Ácido Cítrico/química , Dimerização , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Nanopartículas Metálicas/toxicidade , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Suínos
11.
Nat Nanotechnol ; 16(10): 1150-1160, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34354264

RESUMO

Although nanomaterials have shown promising biomedical application potential, incomplete understanding of their molecular interactions with biological systems prevents their inclusion into mainstream clinical applications. Here we show that black phosphorus (BP) nanomaterials directly affect the cell cycle's centrosome machinery. BP destabilizes mitotic centrosomes by attenuating the cohesion of pericentriolar material and consequently leads to centrosome fragmentation within mitosis. As a result, BP-treated cells exhibit multipolar spindles and mitotic delay, and ultimately undergo apoptosis. Mechanistically, BP compromises centrosome integrity by deactivating the centrosome kinase polo-like kinase 1 (PLK1). BP directly binds to PLK1, inducing its aggregation, decreasing its cytosolic mobility and eventually restricting its recruitment to centrosomes for activation. With this mechanism, BP nanomaterials show great anticancer potential in tumour xenografted mice. Together, our study reveals a molecular mechanism for the tumoricidal properties of BP and proposes a direction for biomedical application of nanomaterials by exploring their intrinsic bioactivities.


Assuntos
Proteínas de Ciclo Celular/genética , Centrossomo/efeitos dos fármacos , Nanoestruturas/química , Neoplasias/tratamento farmacológico , Fósforo/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Animais , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/antagonistas & inibidores , Células HeLa , Xenoenxertos , Humanos , Camundongos , Mitose/efeitos dos fármacos , Neoplasias/genética , Neoplasias/patologia , Fósforo/química , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Quinase 1 Polo-Like
12.
ACS Appl Mater Interfaces ; 13(25): 29936-29948, 2021 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-34143617

RESUMO

Alzheimer's disease (AD) is a major cause of dementia characterized by the overexpression of transmembrane amyloid precursor protein and its neurotoxic byproduct amyloid beta (Aß). A small peptide of considerable hydrophobicity, Aß is aggregation prone catalyzed by the presence of cell membranes, among other environmental factors. Accordingly, current AD mitigation strategies often aim at breaking down the Aß-membrane communication, yet no data is available concerning the cohesive interplay of the three key entities of the cell membrane, Aß, and its inhibitor. Using a lipophilic Laurdan dye and confocal fluorescence microscopy, we observed cell membrane perturbation and actin reorganization induced by Aß oligomers but not by Aß monomers or amyloid fibrils. We further revealed recovery of membrane fluidity by ultrasmall MoS2 quantum dots, also shown in this study as a potent inhibitor of Aß amyloid aggregation. Using discrete molecular dynamics simulations, we uncovered the binding of MoS2 and Aß monomers as mediated by hydrophilic interactions between the quantum dots and the peptide N-terminus. In contrast, Aß oligomers and fibrils were surface-coated by the ultrasmall quantum dots in distinct testudo-like, reverse protein-corona formations to prevent their further association with the cell membrane and adverse effects downstream. This study offers a crucial new insight and a viable strategy for regulating the amyloid aggregation and membrane-axis of AD pathology with multifunctional nanomedicine.


Assuntos
Peptídeos beta-Amiloides , Dissulfetos/química , Fluidez de Membrana/fisiologia , Molibdênio/química , Pontos Quânticos/química , 2-Naftilamina/análogos & derivados , 2-Naftilamina/química , Actinas/química , Actinas/metabolismo , Doença de Alzheimer , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Linhagem Celular Tumoral , Membrana Celular/química , Membrana Celular/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Lauratos/química , Microscopia Confocal , Simulação de Dinâmica Molecular , Nanomedicina
13.
Small ; 16(41): e2003000, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32803846

RESUMO

Anti-hypoxia cancer nanomedicine (AHCN) holds exciting potential in improving oxygen-dependent therapeutic efficiencies of malignant tumors. However, most studies regarding AHCN focus on optimizing structure and function of nanomaterials with presupposed successful entry into tumor cells. From such a traditional perspective, the main barrier that AHCN needs to overcome is mainly the tumor cell membrane. However, such an oversimplified perspective would neglect that real tumors have many biological, physiological, physical, and chemical defenses preventing the current state-of-the-art AHCNs from even reaching the targeted tumor cells. Fortunately, in recent years, some studies are beginning to intentionally focus on overcoming physiological barriers to alleviate hypoxia. In this Review, the limitations behind the traditional AHCN delivery mindset are addressed and the key barriers that need to be surmounted before delivery to cancer cells and some good ways to improve cell membrane attachment, internalization, and intracellular retention are summarized. It is aimed to contribute to Review literature on this emerging topic through refreshing perspectives based on this work and what is also learnt from others. This Review would therefore assist AHCNs researchers to have a quick overview of the essential information and glean thought-provoking ideas to advance this sub-field in cancer nanomedicine.


Assuntos
Nanoestruturas , Neoplasias , Sistemas de Liberação de Medicamentos , Humanos , Hipóxia , Nanomedicina , Neoplasias/tratamento farmacológico , Oxigênio/uso terapêutico
14.
Small ; 16(19): e1906975, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32301572

RESUMO

DNA nanostructures as scaffolds for drug delivery, biosensing, and bioimaging are hindered by its vulnerability in physiological settings, less favorable of incorporating arbitrary guest molecules and other desirable functionalities. Noncanonical self-assembly of DNA nanostructures with small molecules in an alternative system is an attractive strategy to expand their applications in multidisciplinary fields and is rarely explored. This work reports a nitrogen-enriched carbon dots (NCDs)-mediated DNA nanostructure self-assembly strategy. Given the excellent photoluminescence and photodynamic properties of NCDs, the obtained DNA/NCDs nanocomplex holds great potential for bioimaging and anticancer therapy. NCDs can mediate DNA nanoprism (NPNCD ) self-assembly isothermally at a large temperature and pH range in a magnesium-free manner. To explore the suitability of NPNCD in potential biomedical applications, the cytotoxicity and cellular uptake efficiency of NPNCD are evaluated. NPNCD with KRAS siRNA (NPNCD K) is further conjugated for KRAS-mutated nonsmall cell lung cancer therapy. The NPNCD K shows excellent gene knockdown efficiency and anticancer effect in vitro. The current study suggests that conjugating NCDs with programmable DNA nanostructures is a powerful strategy to endow DNA nanostructures with new functionalities, and NPNCD may be a potential theranostic platform with further fine-tuned properties of CDs such as near-red fluorescence or photothermal activities.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Nanoestruturas , Carbono , DNA , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Nitrogênio , Medicina de Precisão , Nanomedicina Teranóstica
15.
ACS Nano ; 14(3): 3259-3271, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32049490

RESUMO

The cancer cell membrane contains an arsenal of highly specific homotypic moieties that can be used to recognize its own kind. These cell membranes are often used to coat spherical nanoparticles to enhance nanomedicines' targeting specificities and uptakes. A sphere, however, has only a point contact with a surface at any given time. It is shown here that, by retaining a flatter morphology of the cracked cell membrane through stiffening with in situ synthesized gold nanomaterials, an increased area of interaction could be maintained and hence improve upon the in vitro and in vivo homotypic targeting capabilities between cancer cell types. This enhancement is especially important in vivo as any nanomedicine with targeting moieties probably has a single pass at interacting with the target cell before subsequent system clearance. Possible future clinical applications may involve the usage of a patient's autologous tumor biopsy tissues, which are very limited in supply, and therefore ensuring that we capitalize on the entire collective surface area of the cancer cell membrane available becomes an important consideration in the design and delivery our cell membrane-derived nanomedicines.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Melanoma/tratamento farmacológico , Nanomedicina , Animais , Antibióticos Antineoplásicos/química , Membrana Celular/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Doxorrubicina/química , Ensaios de Seleção de Medicamentos Antitumorais , Ouro/química , Humanos , Melanoma/diagnóstico por imagem , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Nanoestruturas/química , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/tratamento farmacológico , Imagem Óptica , Tamanho da Partícula , Propriedades de Superfície
16.
Nanoscale ; 12(1): 43-57, 2020 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-31799539

RESUMO

Quantum dots (QDs) originating from two-dimensional (2D) sheets of graphitic carbon nitride (g-C3N4), graphene, hexagonal boron nitride (h-BN), monoatomic buckled crystals (phosphorene), germanene, silicene and transition metal dichalcogenides (TMDCs) are emerging zero-dimensional materials. These QDs possess diverse optical properties, are chemically stable, have surprisingly excellent biocompatibility and are relatively amenable to surface modifications. It is therefore not difficult to see that these QDs have potential in a variety of bioapplications, including biosensing, bioimaging and anticancer and antimicrobial therapy. In this review, we briefly summarize the recent progress of these exciting QD based nanoagents and strategies for phototherapy. In addition, we will discuss about the current limitations, challenges and future prospects of QDs in biomedical applications.


Assuntos
Pontos Quânticos/química , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/uso terapêutico , Grafite/química , Humanos , Nanoestruturas/química , Neoplasias/patologia , Neoplasias/terapia , Compostos de Nitrogênio/química , Fósforo/química , Fototerapia , Propriedades de Superfície
17.
Chem Soc Rev ; 48(21): 5381-5407, 2019 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-31495856

RESUMO

The ever-growing use of inorganic nanoparticles (NPs) in biomedicine provides an exciting approach to develop novel imaging and drug delivery systems, owing to the ease with which these NPs can be functionalized to cater to various applications. In cancer therapeutics, nanomedicine generally relies on the enhanced permeability and retention (EPR) effect observed in tumour vasculature to deliver anti-cancer drugs across the endothelium. However, such a phenomenon is dependent on the tumour microenvironment and is not consistently observed in all tumour types, thereby limiting drug transport to the tumour site. On the other hand, there is a rise in utilizing inorganic NPs to intentionally induce endothelial leakiness, creating a window of opportunity to control drug delivery across the endothelium. While this active targeting approach creates a similar phenomenon compared to the EPR effect arising from tumour tissues, its drug delivery applications extend beyond cancer therapeutics and into other vascular-related diseases. In this review, we summarize the current findings of the EPR effect and assess its limitations in the context of anti-cancer drug delivery systems. While the EPR effect offers a possible route for drug passage, we further explore alternative uses of NPs to create controllable endothelial leakiness within short exposures, a phenomenon we coined as nanomaterial-induced endothelial leakiness (NanoEL). Furthermore, we discuss the main mechanistic features of the NanoEL effect that make it unique from conventionally established endothelial leakiness in homeostatic and pathologic conditions, as well as examine its potential applicability in vascular-related diseases, particularly cancer. Therefore, this new paradigm changes the way inorganic NPs are currently being used for biomedical applications.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Nanomedicina , Nanopartículas/química , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Antineoplásicos/química , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Humanos , Neoplasias/patologia , Neovascularização Patológica/patologia
18.
Nanotoxicology ; 13(5): 682-700, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30776942

RESUMO

Nanoparticles (NPs) have been widely used in biomedical field for therapeutic treatments, drug carriers, and bio-imaging agent. Recent studies have highlighted the possibility of utilizing inorganic NPs in inducing endothelial leakiness through endothelial remodeling to promote drug transport across the barrier. However, an uncontrolled and persistent leakiness could lead to promiscuous transport of molecules and cells across the barrier, highlighting the pressing need to control the timely recovery from endothelial cell leakiness. Herein, we show that angiopoietin-1 (Ang1) could promote recovery of human microvascular endothelial cells (HMVECs) from titanium dioxide nanoparticle (TiO2 NPs)-induced endothelial leakiness. Ang1 is known as an anti-permeability growth factor which forms complexes with its receptor Tie2 at the cell-to-cell junctions. We find that the introduction of Ang1 not only accelerates the recovery of NP-induced endothelial leakiness (NanoEL) but also promotes cell rigidity by increasing tubulin acetylation, thereby remodels the endothelial cells to further mitigate the effects of NP exposure through the activation of the Akt pathway. Using in vitro metastasis model, we further show that HMVECs treated with TiO2 NPs followed by Ang1 could reduce migration of human skin cancer A431 cells across the endothelial barrier. In summary, Ang1 plays important roles in promoting the recovery of endothelial cell leakiness and endothelial stability through a mechano-transduction pathway and shows great potential as key modulator that allows material scientist to regulate endothelial leakiness induced by NPs.


Assuntos
Angiopoietina-1/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Microvasos/efeitos dos fármacos , Nanopartículas/toxicidade , Titânio/toxicidade , Angiopoietina-1/genética , Animais , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Microvasos/citologia , Receptor TIE-2/metabolismo
19.
Nat Nanotechnol ; 14(3): 279-286, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30692675

RESUMO

While most cancer nanomedicine is designed to eliminate cancer, the nanomaterial per se can lead to the formation of micrometre-sized gaps in the blood vessel endothelial walls. Nanomaterials-induced endothelial leakiness (NanoEL) might favour intravasation of surviving cancer cells into the surrounding vasculature and subsequently extravasation, accelerating metastasis. Here, we show that nanoparticles induce endothelial leakiness through disruption of the VE-cadherin-VE-cadherin homophilic interactions at the adherens junction. We show that intravenously injected titanium dioxide, silica and gold nanoparticles significantly accelerate both intravasation and extravasation of breast cancer cells in animal models, increasing the extent of existing metastasis and promoting the appearance of new metastatic sites. Our results add to the understanding of the behaviour of nanoparticles in complex biological systems. The potential for NanoEL needs to be taken into consideration when designing future nanomedicines, especially nanomedicine to treat cancer.


Assuntos
Neoplasias da Mama/patologia , Células Endoteliais/patologia , Extravasamento de Materiais Terapêuticos e Diagnósticos/patologia , Nanopartículas Metálicas/química , Animais , Vasos Sanguíneos/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Metástase Neoplásica , Células Neoplásicas Circulantes/patologia , Permeabilidade , Titânio/química
20.
Nat Commun ; 10(1): 41, 2019 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-30604777

RESUMO

Transition metal dichalcogenide (TMD) quantum dots (QDs) are fundamentally interesting because of the stronger quantum size effect with decreased lateral dimensions relative to their larger 2D nanosheet counterparts. However, the preparation of a wide range of TMD QDs is still a continual challenge. Here we demonstrate a bottom-up strategy utilizing TM oxides or chlorides and chalcogen precursors to synthesize a small library of TMD QDs (MoS2, WS2, RuS2, MoTe2, MoSe2, WSe2 and RuSe2). The reaction reaches equilibrium almost instantaneously (~10-20 s) with mild aqueous and room temperature conditions. Tunable defect engineering can be achieved within the same reactions by deviating the precursors' reaction stoichiometries from their fixed molecular stoichiometries. Using MoS2 QDs for proof-of-concept biomedical applications, we show that increasing sulfur defects enhanced oxidative stress generation, through the photodynamic effect, in cancer cells. This facile strategy will motivate future design of TMDs nanomaterials utilizing defect engineering for biomedical applications.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA