Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Biochemistry ; 60(13): 941-955, 2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-32452667

RESUMO

There has been significant interest in developing cell membrane-coated nanoparticles due to their unique abilities of biomimicry and biointerfacing. As the technology progresses, it becomes clear that the application of these nanoparticles can be drastically broadened if additional functions beyond those derived from the natural cell membranes can be integrated. Herein, we summarize the most recent advances in the functionalization of cell membrane-coated nanoparticles. In particular, we focus on emerging methods, including (1) lipid insertion, (2) membrane hybridization, (3) metabolic engineering, and (4) genetic modification. These approaches contribute diverse functions in a nondisruptive fashion while preserving the natural function of the cell membranes. They also improve on the multifunctional and multitasking ability of cell membrane-coated nanoparticles, making them more adaptive to the complexity of biological systems. We hope that these approaches will serve as inspiration for more strategies and innovations to advance cell membrane coating technology.


Assuntos
Membrana Celular/química , Nanopartículas/química , Nanotecnologia/métodos
2.
J Am Chem Soc ; 143(42): 17615-17621, 2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34647745

RESUMO

Cellular binding and entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are mediated by its spike glycoprotein (S protein), which binds with not only the human angiotensin-converting enzyme 2 (ACE2) receptor but also glycosaminoglycans such as heparin. Cell membrane-coated nanoparticles ("cellular nanosponges") mimic the host cells to attract and neutralize SARS-CoV-2 through natural cellular receptors, leading to a broad-spectrum antiviral strategy. Herein, we show that increasing surface heparin density on the cellular nanosponges can promote their inhibition against SARS-CoV-2. Specifically, cellular nanosponges are made with azido-expressing host cell membranes followed by conjugating heparin to the nanosponge surfaces. Cellular nanosponges with a higher heparin density have a larger binding capacity with viral S proteins and a significantly higher inhibition efficacy against SARS-CoV-2 infectivity. Overall, surface glycan engineering of host-mimicking cellular nanosponges is a facile method to enhance SARS-CoV-2 inhibition. This approach can be readily generalized to promote the inhibition of other glycan-dependent viruses.


Assuntos
Tratamento Farmacológico da COVID-19 , Heparina/administração & dosagem , Nanoestruturas/uso terapêutico , Polissacarídeos/administração & dosagem , SARS-CoV-2/metabolismo , COVID-19/virologia , Heparina/metabolismo , Humanos , Polissacarídeos/metabolismo
3.
Nano Lett ; 18(2): 1498-1505, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29342359

RESUMO

Nanomedicine have shown success in cancer therapy, but the pharmacological actions of most nanomedicine are often nonspecific to cancer cells because of utilization of the therapeutic agents that induce cell apoptosis from inner organelles. We herein report the development of semiconducting photothermal nanoagonists that can remotely and specifically initiate the apoptosis of cancer cells from cell membrane. The organic nanoagonists comprise semiconducting polymer nanoparticles (SPNs) and capsaicin (Cap) as the photothermally responsive nanocarrier and the agonist for activation of transient receptor potential cation channel subfamily V member 1 (TRPV1), respectively. Under multiple NIR laser irradiation at the time scale of seconds, the nanoagonists can repeatedly and locally release Cap to multiply activate TRPV1 channels on the cellular membrane; the cumulative effect is the overinflux of ions in mitochondria followed by the induction of cell apoptosis specifically for TRPV1-postive cancer cells. Multiple transient activation of TRPV1 channels is essential to induce such a cell death both in vitro and in vivo because both free Cap and simple Cap-encapsulated nanoparticles fail to do so. The photothermally triggered release also ensures a high local concentration of the TRPV1 agonist at tumor site, permitting specific cancer cell therapy at a low systemic administration dosage. Our study thus demonstrates the first example of ion-channel-specific and remote-controlled drug-delivery system for cancer cell therapy.


Assuntos
Capsaicina/administração & dosagem , Preparações de Ação Retardada/química , Neoplasias/tratamento farmacológico , Pontos Quânticos/química , Canais de Cátion TRPV/agonistas , Animais , Capsaicina/uso terapêutico , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Células HeLa , Humanos , Raios Infravermelhos , Camundongos , Células NIH 3T3 , Canais de Cátion TRPV/metabolismo , Temperatura
4.
Bioconjug Chem ; 29(4): 928-938, 2018 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-29466856

RESUMO

Near-infrared (NIR) light-mediated photodynamic therapy (PDT), especially based on lanthanide-doped upconversion nanocrystals (UCNs), have been extensively investigated as a promising strategy for effective cellular ablation owing to their unique optical properties to convert NIR light excitation into multiple short-wavelength emissions. Despite the deep tissue penetration of NIR light in living systems, the therapeutic efficiency is greatly restricted by insufficient oxygen supply in hypoxic tumor microenvironment. Moreover, the coexistent tumor-associated macrophages (TAMs) play critical roles in tumor recurrence during the post-PDT period. Herein, we developed a unique photosensitizer-loaded UCNs nanoconjugate (PUN) by integrating manganese dioxide (MnO2) nanosheets and hyaluronic acid (HA) biopolymer to improve NIR light-mediated PDT efficacy through attenuating hypoxia status and synergistically reprogramming TAMs populations. After the reaction with overproduced H2O2 in acidic tumor microenvironment, the MnO2 nanosheets were degraded for the production of massive oxygen to greatly enhance the oxygen-dependent PDT efficiency upon 808 nm NIR light irradiation. More importantly, the bioinspired polymer HA could effectively reprogram the polarization of pro-tumor M2-type TAMs to anti-tumor M1-type macrophages to prevent tumor relapse after PDT treatment. Such promising results provided the great opportunities to achieve enhanced cellular ablation upon NIR light-mediated PDT treatment by attenuating hypoxic tumor microenvironment, and thus facilitated the rational design of new generations of nanoplatforms toward immunotherapy to inhibit tumor recurrence during post-PDT period.


Assuntos
Hipóxia/tratamento farmacológico , Macrófagos/efeitos dos fármacos , Compostos de Manganês/uso terapêutico , Melanoma/tratamento farmacológico , Nanopartículas/uso terapêutico , Óxidos/uso terapêutico , Fármacos Fotossensibilizantes/uso terapêutico , Microambiente Tumoral/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Ácido Hialurônico/química , Ácido Hialurônico/uso terapêutico , Hipóxia/metabolismo , Hipóxia/patologia , Raios Infravermelhos , Elementos da Série dos Lantanídeos/química , Elementos da Série dos Lantanídeos/uso terapêutico , Macrófagos/metabolismo , Macrófagos/patologia , Compostos de Manganês/química , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Nanopartículas/química , Óxidos/química , Oxigênio/metabolismo , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/química , Células RAW 264.7
5.
Bioconjug Chem ; 29(4): 838-851, 2018 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-29509403

RESUMO

In terms of the extremely small size and large specific surface area, nanomaterials often exhibit unusual physical and chemical properties, which have recently attracted considerable attention in bionanotechnology and nanomedicine. Currently, the extensive usage of nanotechnology in medicine holds great potential for precise diagnosis and effective therapeutics of various human diseases in clinical practice. However, a detailed understanding regarding how nanomedicine interacts with the intricate environment in complex living systems remains a pressing and challenging goal. Inspired by the diversified membrane structures and functions of natural prototypes, research activities on biomimetic and bioinspired membranes, especially for those cloaking nanosized platforms, have increased exponentially. By taking advantage of the flexible synthesis and multiple functionality of nanomaterials, a variety of unique nanostructures including inorganic nanocrystals and organic polymers have been widely devised to substantially integrate with intrinsic biomoieties such as lipids, glycans, and even cell and bacteria membrane components, which endow these abiotic nanomaterials with specific biological functionalities for the purpose of detailed investigation of the complicated interactions and activities of nanomedicine in living bodies, including their immune response activation, phagocytosis escape, and subsequent clearance from vascular system. In this review, we summarize the strategies established recently for the development of biomimetic membrane-cloaked nanoplatforms derived from inherent host cells (e.g., erythrocytes, leukocytes, platelets, and exosomes) and invasive pathogens (e.g., bacteria and viruses), mainly attributed to their versatile membrane properties in biological fluids. Meanwhile, the promising biomedical applications based on nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective vaccine development for disease prevention, have also been outlined. Finally, the potential challenges and future prospects of the biomimetic membrane-cloaked nanoplatforms are also discussed.


Assuntos
Materiais Biomiméticos/química , Nanomedicina/métodos , Nanoestruturas/química , Animais , Bactérias/química , Materiais Biomiméticos/uso terapêutico , Biomimética/métodos , Plaquetas/química , Membrana Celular/química , Sistemas de Liberação de Medicamentos/métodos , Eritrócitos/química , Exossomos/química , Humanos , Leucócitos/química , Nanoestruturas/uso terapêutico , Nanotecnologia/métodos , Vírus/química
6.
Angew Chem Int Ed Engl ; 56(11): 3031-3035, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28157258

RESUMO

The spatiotemporal regulation of light-gated ion channels is a powerful tool to study physiological pathways and develop personalized theranostic modalities. So far, most existing light-gated channels are limited by their action spectra in the ultraviolet (UV) or visible region. Simple and innovative strategies for the specific attachment of photoswitches on the cell surface without modifying or genetically encoding channel structures, and more importantly, that enable the remote activation of ion-channel functions within near-infrared (NIR) spectral window in living systems, remain a challenging concern. Herein, metabolic glycan biosynthesis is used to achieve site-specific covalent attachment of near-infrared-light-mediated lanthanide-doped upconversion nanocrystals (UCNs) to the cell surface through copper-free click cyclization. Upon irradiation with 808 nm light, the converted emission at 480 nm could activate a light-gated ion channel, channelrhodopsins-2 (ChR2), and thus remotely control the cation influx. This unique strategy provides valuable insights on the specific regulation membrane-associated activities in vivo.

7.
Anal Biochem ; 438(1): 22-8, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23499968

RESUMO

In this article, a kind of simple, sensitive, and rapid quantum dots (QDs)-based multiplex DNA sensor is developed for the simultaneous detection of BRAF and BRCA DNA based on the "nano-on-micro" technique. In our strategy, capture DNA(BRCA) and DNA(BRAF) are simultaneously immobilized on the surface of amino-modified silica microbeads. After blocking with bovine serum albumin (BSA), different concentrations of target DNA(BRCA) and DNA(BRAF) are introduced to hybrid with complementary capture DNA(BRCA) and DNA(BRAF). After hybridization, QDs546-labeled probe DNA(BRAF) and QDs657-labeled probe DNA(BRCA) were added into the above solution so that the unreacted capture DNA(BRCA) and DNA(BRAF) could be detected by QDs657-labeled probe DNA(BRCA) and QDs546-labeled probe DNA(BRAF) simultaneously. We demonstrate that the proposed method is effective for detecting BRAF and BRCA DNA with high sensitivity. The sensor has great potential to expand its application to the early diagnosis of cancers such as breast cancer, ovarian cancer, and papillary thyroid carcinoma.


Assuntos
Técnicas Biossensoriais/métodos , DNA/análise , Proteínas Proto-Oncogênicas B-raf/genética , Pontos Quânticos , Proteínas Supressoras de Tumor/genética , DNA/sangue , DNA/química , Humanos , Concentração de Íons de Hidrogênio , Microtecnologia , Nanotecnologia , Hibridização de Ácido Nucleico , Concentração Osmolar , Espectrometria de Fluorescência
8.
Adv Healthc Mater ; 12(23): e2300351, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37289567

RESUMO

Vaccines provide a powerful tool to modulate the immune system for human disease prevention and treatment. Classical vaccines mainly initiate immune responses in the lymph nodes (LNs) after subcutaneous injection. However, some vaccines suffer from inefficient delivery of antigens to LNs, undesired inflammation, and slow immune induction when encountering the rapid proliferation of tumors. Alternatively, the spleen, as the largest secondary lymphoid organ with a high density of antigen-presenting cells (APCs) and lymphocytes, acts as an emerging target organ for vaccinations in the body. Upon intravenous administration, the rationally designed spleen-targeting nanovaccines can be internalized by the APCs in the spleen to induce selective antigen presentation to T and B cells in their specific sub-regions, thereby rapidly boosting durable cellular and humoral immunity. Herein, the recent advances of spleen-targeting nanovaccines for immunotherapy based on the anatomical architectures and functional zones of the spleen, as well as their limitations and perspectives for clinical applications are systematically summarized. The aim is to emphasize the design of innovative nanovaccines for enhanced immunotherapy of intractable diseases in the future.


Assuntos
Vacinas Anticâncer , Neoplasias , Vacinas , Humanos , Baço , Antígenos , Apresentação de Antígeno , Imunoterapia
9.
Biomaterials ; 302: 122330, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37742508

RESUMO

Botulinum toxin (BoNT) is a potent neurotoxin that poses a significant threat as a biowarfare weapon and a potential bioterrorist tool. Currently, there is a lack of effective countermeasures to combat BoNT intoxication in the event of a biological attack. Here, we report on a novel solution by combining cell metabolic engineering with cell membrane coating nanotechnology, resulting in the development of glycan-modified cellular nanosponges that serve as a biomimetic and broad-spectrum BoNT detoxification strategy. Specifically, we increase the expression levels of gangliosides on THP-1 cells through metabolic engineering, and then collect the modified THP-1 cell membrane and coat it onto synthetic polymeric cores, creating cellular nanosponges that closely mimic host cells. Our findings demonstrate that higher levels of gangliosides on the cellular nanosponges result in greater binding capacities with BoNT. The glycan-modified cellular nanosponges exhibit superior efficacy in neutralizing BoNT cytotoxicity in vitro when compared to their unmodified counterparts. In a mouse model of BoNT intoxication, the glycan-modified cellular nanosponges show more pronounced survival benefits when administered both as a treatment and a preventative regimen. These results highlight the potential of cellular nanosponges, especially when modified with glycans, as a promising countermeasure platform against BoNT and related clostridial toxins.


Assuntos
Toxinas Botulínicas , Camundongos , Animais , Membrana Celular/metabolismo , Gangliosídeos/metabolismo , Polissacarídeos
10.
ACS Nano ; 16(11): 19145-19154, 2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36354967

RESUMO

Neurotoxins attack and destruct the nervous system, which can cause serious health problems and security threats. Existing detoxification approaches, such as antibodies and small molecule antidotes, rely on neurotoxin's molecular structure as design cues and require toxin-specific development for each type of toxins. However, the enormous diversity of neurotoxins makes such structure-based development of antitoxin particularly challenging and inefficient. Here, we report on the development and use of neuronal membrane-coated nanosponges (denoted "Neuron-NS") as an effective approach to detoxifying neurotoxins. Specifically, Neuron-NS act as neuron decoys to lure neurotoxins, bind with and neutralize the toxins, and thus block them from attacking the host neuron cells. These nanosponges detoxify neurotoxins regardless of their molecular structures and therefore can overcome the challenge posed by toxin structural diversity. In the study, we fabricate Neuron-NS by coating the membrane of Neuro-2a cells onto polymeric cores. Meanwhile, we select tetrodotoxin (TTX) as a model neurotoxin and demonstrate the detoxification efficacy of the Neuron-NS in a cytotoxicity assay, a calcium flux assay, and a cell osmotic swelling assay in vitro. Additionally, in mouse models of TTX intoxication, the Neuron-NS significantly enhance mouse survival in therapeutic and prophylactic regimens without showing acute toxicity. Overall, the Neuron-NS contribute to the current detoxification arsenal with the potential to treat various injuries and diseases caused by neurotoxins.


Assuntos
Neurônios , Neurotoxinas , Camundongos , Animais , Neurotoxinas/toxicidade , Neurotoxinas/química , Antídotos
11.
Adv Healthc Mater ; 11(7): e2101349, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34468090

RESUMO

White blood cells (WBCs) are immune cells that play essential roles in critical diseases including cancers, infections, and inflammatory disorders. Their dynamic and diverse functions have inspired the development of WBC membrane-coated nanoparticles (denoted "WBC-NPs"), which are formed by fusing the plasma membranes of WBCs, such as macrophages, neutrophils, T cells, and natural killer cells, onto synthetic nanoparticle cores. Inheriting the entire source cell antigens, WBC-NPs act as source cell decoys and simulate their broad biointerfacing properties with intriguing therapeutic potentials. Herein, the recent development and medical applications of WBC-NPs focusing on four areas, including WBC-NPs as carriers for drug delivery, as countermeasures for biological neutralization, as nanovaccines for immune modulation, and as tools for the isolation of circulating tumor cells and fundamental research is reviewed. Overall, the recent development and studies of WBC-NPs have established the platform as versatile nanotherapeutics and tools with broad medical application potentials.


Assuntos
Nanopartículas , Neoplasias , Membrana Celular/metabolismo , Sistemas de Liberação de Medicamentos , Humanos , Leucócitos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo
12.
Bioeng Transl Med ; 6(1): e10187, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33532587

RESUMO

Current drug delivery approaches for the treatment of cartilage disorders such as osteoarthritis (OA) remain inadequate to achieve sufficient drug penetration and retention in the dense cartilage matrix. Herein, we synthesize sub-30 nm lipid-polymer hybrid nanoparticles functionalized with collagen-targeting peptides for targeted drug delivery to the cartilage. The nanoparticles consist of a polymeric core for drug encapsulation and a lipid shell modified with a collagen-binding peptide. By combining these design features, the nanoparticles can penetrate deep and accumulate preferentially in the cartilage. Using MK-8722, an activator of 5'-adenosine monophosphate-activated protein kinase (AMPK), as a model drug, the nanoparticles can encapsulate the drug molecules in high capacity and release them in a sustained and controllable manner. When injected into the knee joints of the mice with collagenase-induced OA, the drug-loaded nanoparticles can effectively reduce cartilage damage and alleviate the disease severity. Overall, the ultrasmall targeted nanoparticles represent a promising delivery platform to overcome barriers of dense tissues for the treatment of various indications, including cartilage disorders.

13.
Adv Biosyst ; 3(1): e1800233, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32627341

RESUMO

Membrane ion channels are ultimately responsible for the propagation and integration of electrical signals in the nervous, muscular, and other systems. Their activation or malfunctioning plays a significant role in physiological and pathophysiological processes. Using optogenetics to dynamically and spatiotemporally control ion channels has recently attracted considerable attention. However, most of the established optogenetic tools (e.g., channelrhodopsins, ChRs) for optical manipulations, are mainly stimulated by UV or visible light, which raises the concerns of potential photodamage, limited tissue penetration, and high-invasive implantation of optical fiber devices. Near-infrared (NIR) upconversion nanoparticle (UCNP)-mediated optogenetic systems provide great opportunities for overcoming the problems encountered in the manipulation of ion channels in deep tissues. Hence, this review focuses on the recent advances in NIR regulation of membrane ion channels via upconversion optogenetics in biomedical research. The engineering and applications of upconversion optogenetic systems by the incorporation multiple emissive UCNPs into various light-gated ChRs/ligands are first elaborated, followed by a detailed discussion of the technical improvements for more precise and efficient control of membrane channels. Finally, the future perspectives for refining and advancing NIR-mediated upconversion optogenetics into in vivo even in clinical applications are proposed.

14.
Nat Commun ; 10(1): 1087, 2019 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-30842426

RESUMO

Precise and differential profiling of the dynamic correlations and pathophysiological implications of multiplex biological mediators with deep penetration and highly programmed precision remain critical challenges in clinics. Here we present an innovative strategy by tailoring a powerful multispectral optoacoustic tomography (MSOT) technique with a photon-upconverting nanoprobe (UCN) for simultaneous visualization of diversely endogenous redox biomarkers with excellent spatiotemporal resolution in living conditions. Upon incorporating two specific radicals-sensitive NIR cyanine fluorophores onto UCNs surface, such nanoprobes can orthogonally respond to disparate oxidative and nitrosative stimulation, and generate spectrally opposite optoacoustic signal variations, which thus achieves compelling superiorities for reversed ratiometric tracking of multiple radicals under dual independent wavelength channels, and significantly, for precise validating of their complex dynamics and correlations with redox-mediated pathophysiological procession in vivo.


Assuntos
Inflamação/diagnóstico por imagem , Sondas Moleculares/química , Nanopartículas/química , Técnicas Fotoacústicas/métodos , Fótons , Animais , Biomarcadores/análise , Modelos Animais de Doenças , Feminino , Corantes Fluorescentes/administração & dosagem , Corantes Fluorescentes/química , Coração/diagnóstico por imagem , Humanos , Inflamação/imunologia , Inflamação/patologia , Rim/diagnóstico por imagem , Rim/patologia , Fígado/diagnóstico por imagem , Fígado/patologia , Pulmão/diagnóstico por imagem , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Sondas Moleculares/administração & dosagem , Nanopartículas/administração & dosagem , Oxirredução , Células RAW 264.7 , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Baço/diagnóstico por imagem , Baço/patologia , Nanomedicina Teranóstica/métodos , Tomografia Computadorizada por Raios X/métodos
15.
Chem Sci ; 11(3): 803-811, 2019 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-34123056

RESUMO

Pharmacological interventions for effective treatment require opportune, dynamic and accurate manifestation of pathological status. Traditional clinical techniques relying on biopsy-based histological examinations and blood tests are dramatically restricted due to their invasiveness, unsatisfactory precision, non-real-time reporting and risk of complications. Although current strategies through molecular imaging enable non-invasive and spatiotemporal mapping of pathological changes in intact organisms, environment-activatable, sensitive and quantitative sensing platforms, especially for dynamic feedback of the therapeutic response, are still urgently desired in practice. Herein, we innovatively integrate deep-tissue penetrable multispectral optoacoustic tomography (MSOT) and near-infrared (NIR) optical imaging based technology by tailoring a free radical-responsive chromophore with photon-upconverting nanocrystals. During the therapeutic process, the specific reactions between the drug-stimulated reactive oxygen species (ROS) and radical-sensitive probes result in an absorption shift, which can be captured by MSOT. Meanwhile, the radical-triggered reaction also induces multispectral upconversion luminescence (UCL) responses that exhibit the opposite trend in comparison to MSOT. Such reversed-ratiometric dual-modal imaging outcomes provide an ideal cross-referencing system that guarantees the maximum sensing specificity and sensitivity, thus enabling precise disease biology evaluation and treatment assessments in vivo.

16.
J Vis Exp ; (129)2017 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-29155787

RESUMO

Lanthanide-doped upconversion nanocrystals (UCNs) have attracted much attention in recent years based on their promising and controllable optical properties, which allow for the absorption of near-infrared (NIR) light and can subsequently convert it into multiplexed emissions that span over a broad range of regions from the UV to the visible to the NIR. This article presents detailed experimental procedures for high-temperature co-precipitation synthesis of core-shell UCNs that incorporate different lanthanide ions into nanocrystals for efficiently converting deep-tissue penetrable NIR excitation (808 nm) into a strong blue emission at 480 nm. By controlling the surface modification with biocompatible polymer (polyacrylic acid, PAA), the as-prepared UCNs acquires great solubility in buffer solutions. The hydrophilic nanocrystals are further functionalized with specific ligands (dibenzyl cyclooctyne, DBCO) for localization on the cell membrane. Upon NIR light (808 nm) irradiation, the upconverted blue emission can effectively activate the light-gated channel protein on the cell membrane and specifically regulate the cation (e.g., Ca2+) influx in the cytoplasm. This protocol provides a feasible methodology for the synthesis of core-shell lanthanide-doped UCNs and subsequent biocompatible surface modification for further cellular applications.


Assuntos
Interações Hidrofóbicas e Hidrofílicas , Elementos da Série dos Lantanídeos/síntese química , Nanopartículas/química , Elementos da Série dos Lantanídeos/química , Modelos Moleculares
17.
Theranostics ; 6(13): 2439-2457, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27877246

RESUMO

Currently, precision theranostics have been extensively demanded for the effective treatment of various human diseases. Currently, efficient therapy at the targeted disease areas still remains challenging since most available drug molecules lack of selectivity to the pathological sites. Among different approaches, light-mediated therapeutic strategy has recently emerged as a promising and powerful tool to precisely control the activation of therapeutic reagents and imaging probes in vitro and in vivo, mostly attributed to its unique properties including minimally invasive capability and highly spatiotemporal resolution. Although it has achieved initial success, the conventional strategies for light-mediated theranostics are mostly based on the light with short wavelength (e.g., UV or visible light), which may usually suffer from several undesired drawbacks, such as limited tissue penetration depth, unavoidable light absorption/scattering and potential phototoxicity to healthy tissues, etc. Therefore, a near-infrared (NIR) light-mediated approach on the basis of long-wavelength light (700-1000 nm) irradiation, which displays deep-tissue penetration, minimized photo-damage and low autofluoresence in living systems, has been proposed as an inspiring alternative for precisely phototherapeutic applications in the last decades. Despite numerous NIR light-responsive molecules have been currently proposed for clinical applications, several inherent drawbacks, such as troublesome synthetic procedures, low water solubility and limited accumulation abilities in targeted areas, heavily restrict their applications in deep-tissue therapeutic and imaging studies. Thanks to the amazing properties of several nanomaterials with large extinction coefficient in the NIR region, the construction of NIR light responsive nanoplatforms with multifunctions have become promising approaches for deep-seated diseases diagnosis and therapy. In this review, we summarized various light-triggered theranostic strategies and introduced their great advances in biomedical applications in recent years. Moreover, some other promising light-assisted techniques, such as photoacoustic and Cerenkov radiation, were also systemically discussed. Finally, the potential challenges and future perspectives for light-mediated deep-tissue diagnosis and therapeutics were proposed.


Assuntos
Fototerapia/métodos , Nanomedicina Teranóstica/métodos , Humanos , Raios Infravermelhos/uso terapêutico , Terapia de Alvo Molecular/métodos , Terapia de Alvo Molecular/tendências , Fototerapia/tendências , Nanomedicina Teranóstica/tendências
18.
ACS Appl Mater Interfaces ; 8(29): 18732-40, 2016 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-27367152

RESUMO

In this work, pH/redox dual-responsive carbon dots (CDs-RGD-Pt(IV)-PEG) were fabricated for tumor extracellular microenvironment triggered targeting and enhanced anticancer drug delivery. The system consists of fluorescent carbon dots as imaging-guided drug nanocarriers, cisplatin(IV) as prodrug, and RGD peptide as active targeting ligand, which is covered by monomethoxypolyethylene glycol (mPEG) through tumor extracellular pH (6.5-6.8) responsive benzoic-imine bond. The drug nanocarriers could be tracked by multicolor fluorescence of carbon dots. After the hydrolysis of benzoic-imine bond at the tumor extracellular pH to expose the inner targeting RGD peptide, the drug nanocarriers showed effective uptake by cancer cells through RGD-integrin αvß3 (ligand-receptor) interaction. Upon the internalization, the loaded cisplatin(IV) prodrug was reduced to cytotoxic cisplatin in reductive cytosol of cancer cells to exhibit therapeutic effects. Confocal imaging, flow cytometry, and cell viability assays using CDs-RGD-Pt(IV)-PEG were performed to reveal the enhanced uptake and better therapeutic efficiency to cancer cells with high integrin αvß3 expression at tumor extracellular pH than that in physiological condition. The developed CDs-RGD-Pt(IV)-PEG offers a new strategy to provide safe and effective therapeutic agents based on carbon dots for promising cancer therapy.


Assuntos
Carbono/química , Antineoplásicos , Linhagem Celular Tumoral , Cisplatino , Sistemas de Liberação de Medicamentos , Humanos , Integrina alfaVbeta3 , Microambiente Tumoral
19.
ACS Nano ; 10(4): 4410-20, 2016 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-26997431

RESUMO

Carbon dots (CDs) are remarkable nanocarriers due to their promising optical and biocompatible capabilities. However, their practical applicability in cancer therapeutics is limited by their insensitive surface properties to complicated tumor microenvironment in vivo. Herein, a tumor extracellular microenvironment-responsive drug nanocarrier based on cisplatin(IV) prodrug-loaded charge-convertible CDs (CDs-Pt(IV)@PEG-(PAH/DMMA)) was developed for imaging-guided drug delivery. An anionic polymer with dimethylmaleic acid (PEG-(PAH/DMMA)) on the fabricated CDs-Pt(IV)@PEG-(PAH/DMMA) could undergo intriguing charge conversion to a cationic polymer in mildly acidic tumor extracellular microenvironment (pH ∼ 6.8), leading to strong electrostatic repulsion and release of positive CDs-Pt(IV). Importantly, positively charged nanocarrier displays high affinity to negatively charged cancer cell membrane, which results in enhanced internalization and effective activation of cisplatin(IV) prodrug in the reductive cytosol. The in vitro experimental results confirmed that this promising charge-convertible nanocarrier possesses better therapeutic efficiency under tumor extracellular microenvironment than normal physiological condition and noncharge-convertible nanocarrier. The in vivo experiments further demonstrated high tumor-inhibition efficacy and low side effects of the charge-convertible CDs, proving its capability as a smart drug nanocarrier with enhanced therapeutic effects. The present work provides a strategy to promote potential clinical application of CDs in the cancer treatment.


Assuntos
Antineoplásicos/química , Carbono/química , Cisplatino/química , Nanopartículas/química , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/tratamento farmacológico , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular , Cisplatino/efeitos adversos , Cisplatino/farmacologia , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Estabilidade de Medicamentos , Feminino , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Transplante de Neoplasias , Imagem Óptica , Tamanho da Partícula , Poliaminas/química , Polietilenoglicóis/química , Propriedades de Superfície , Distribuição Tecidual
20.
Dalton Trans ; 45(36): 14101-14108, 2016 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-27711690

RESUMO

Platinum-based chemotherapy, although it has been well proven to be effective in the battle against cancer, suffers from limited specificity, severe side effects and drug resistance. The development of new alternatives with potent anticancer effects and improved specificity is therefore urgently needed. Recently, there are some new chemotherapy reagents based on photoactive Re(i) complexes which have been reported as promising alternatives to improve specificity mainly attributed to the spatial and temporal activation process by light irradiation. However, most of them respond to short-wavelength light (e.g. UV, blue or green light), which may cause unwanted photo damage to cells. Herein, we demonstrate a system for near-infrared (NIR) light controlled activation of Re(i) complex cytotoxicity by integration of photoactivatable Re(i) complexes and lanthanide-doped upconversion nanoparticles (UCNPs). Upon NIR irradiation at 980 nm, the Re(i) complex can be locally activated by upconverted UV light emitted from UCNPs and subsequently leads to enhanced cell lethality. Cytotoxicity studies showed effective inactivation of both drug susceptible human ovarian carcinoma A2780 cells and cisplatin resistant subline A2780cis cells by our UCNP based system with NIR irradiation, and there was minimum light toxicity observed in the whole process, suggesting that such a system could provide a promising strategy to control localized activation of Re(i) complexes and therefore minimize potential side effects.


Assuntos
Complexos de Coordenação , Luz , Nanopartículas Metálicas , Rênio , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Quitosana/administração & dosagem , Quitosana/química , Quitosana/efeitos da radiação , Complexos de Coordenação/administração & dosagem , Complexos de Coordenação/química , Complexos de Coordenação/efeitos da radiação , Fluoretos/administração & dosagem , Fluoretos/química , Fluoretos/efeitos da radiação , Humanos , Nanopartículas Metálicas/administração & dosagem , Nanopartículas Metálicas/química , Nanopartículas Metálicas/efeitos da radiação , Rênio/administração & dosagem , Rênio/química , Rênio/efeitos da radiação , Túlio/administração & dosagem , Túlio/química , Túlio/efeitos da radiação , Itérbio/administração & dosagem , Itérbio/química , Itérbio/efeitos da radiação , Ítrio/administração & dosagem , Ítrio/química , Ítrio/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA