Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 182
Filtrar
1.
Chem Asian J ; : e202400268, 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38578217

RESUMO

Photodynamic therapy (PDT) as an emerging therapeutic method has drawn much attention in the treatment field for cancer. Photosensitizer, which can convert photon energy into cytotoxic species under light irradiation, is the core component in PDT. The design of photosensitizers still faces problems of light absorption, targeting, penetration and oxygen dependence. With the rapid progress of material science, various photosensitizers have been developed to produce cytotoxic species for treatment of tumor with high selectivity, safety, and noninvasiveness. Besides, the applications of photosensitizers have been expanded to diverse cancer treatments such as drug release, optogenetics and immune checkpoint blockade. In this review, we summarize the recent advances of photosensitizers in various therapeutic methods for cancer. Prevailing challenges and further prospects associated with photosensitizers are also discussed.

2.
ACS Appl Bio Mater ; 7(4): 2070-2085, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38489843

RESUMO

Retinal diseases, such as age-related macular degeneration, diabetic retinopathy, and retinoblastoma, stand as the leading causes of irreversible vision impairment and blindness worldwide. Effectively administering drugs for retinal diseases poses a formidable challenge due to the presence of complex ocular barriers and elimination mechanisms. Over time, various approaches have been developed to fabricate drug delivery systems for improving retinal therapy including virus vectors, lipid nanoparticles, and polymers. However, conventional nanocarriers encounter issues related to the controllability, efficiency, and safety in the retina. Therefore, the development of smart nanocarriers for effective or more invasive long-term treatment remains a desirable goal. Recently, approaches have surfaced for the intelligent design of nanocarriers, leveraging specific responses to external or internal triggers and enabling multiple functions for retinal therapy such as topical administration, prolonged drug release, and site-specific drug delivery. This Review provides an overview of prevalent retinal pathologies and related pharmacotherapies to enhance the understanding of retinal diseases. It also surveys recent developments and strategies employed in the intelligent design of nanocarriers for retinal disease. Finally, the challenges of smart nanocarriers in potential clinical retinal therapeutic applications are discussed to inspire the next generation of smart nanocarriers.


Assuntos
Doenças Retinianas , Humanos , Doenças Retinianas/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Preparações Farmacêuticas
3.
Bioact Mater ; 36: 330-357, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38496036

RESUMO

Nanovaccines have gathered significant attention for their potential to elicit tumor-specific immunological responses. Despite notable progress in tumor immunotherapy, nanovaccines still encounter considerable challenges such as low delivery efficiency, limited targeting ability, and suboptimal efficacy. With an aim of addressing these issues, engineering customized nanovaccines through modification or functionalization has emerged as a promising approach. These tailored nanovaccines not only enhance antigen presentation, but also effectively modulate immunosuppression within the tumor microenvironment. Specifically, they are distinguished by their diverse sizes, shapes, charges, structures, and unique physicochemical properties, along with targeting ligands. These features of nanovaccines facilitate lymph node accumulation and activation/regulation of immune cells. This overview of bespoke nanovaccines underscores their potential in both prophylactic and therapeutic applications, offering insights into their future development and role in cancer immunotherapy.

4.
ACS Appl Mater Interfaces ; 16(7): 8538-8553, 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38343191

RESUMO

Large osseous void, postsurgical neoplastic recurrence, and slow bone-cartilage repair rate raise an imperative need to develop functional scaffold in clinical osteosarcoma treatment. Herein, a bionic bilayer scaffold constituting croconaine dye-polyethylene glycol@sodium alginate hydrogel and poly(l-lactide)/hydroxyapatite polymer matrix is fabricated to simultaneously achieve a highly efficient killing of osteosarcoma and an accelerated osteochondral regeneration. First, biomimetic osteochondral structure along with adequate interfacial interaction of the bilayer scaffold provide a structural reinforcement for transverse osseointegration and osteochondral regeneration, as evidenced by upregulated specific expressions of collagen type-I, osteopontin, and runt-related transcription factor 2. Meanwhile, thermal ablation of the synthesized nanoparticles and mitochondrial dysfunction caused by continuously released hydroxyapatite induce residual tumor necrosis synergistically. To validate the capabilities of inhibiting tumor growth and promoting osteochondral regeneration of our proposed scaffold, a novel orthotopic osteosarcoma model simulating clinical treatment scenarios of bone tumors is established on rats. Based on amounts of in vitro and in vivo results, an effective killing of osteosarcoma and a suitable osteal-microenvironment modulation of such bionic bilayer composite scaffold are achieved, which provides insightful implications for photonic hyperthermia therapy against osteosarcoma and following osseous tissue regeneration.


Assuntos
Hipertermia Induzida , Osteossarcoma , Ratos , Animais , Alicerces Teciduais/química , Biônica , Materiais Biocompatíveis/química , Durapatita/química , Regeneração Óssea , Osteossarcoma/terapia , Microambiente Tumoral
5.
Anal Chem ; 96(8): 3462-3469, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38358853

RESUMO

It remains a challenge to use a single probe to simultaneously detect extracellular pH fluctuations and specifically recognize cancer cells for precise drug delivery. Here, we engineered a tetrahedral framework nucleic acid-based logic nanoprobe (isgc8-tFNA) on live cell membranes for simultaneously monitoring extracellular pH and targeted drug delivery. Isgc8-tFNA was anchored stably on the cell surface through three cholesterol molecules inserting into the bilayer of the cell membrane. Once responding to the acidic tumor microenvironment, isgc8-tFNA formed an i-motif structure, leading to turn-on FRET signals for monitoring changes of extracellular pH. The nanoprobe exhibited a narrow pH-response window and excellent reversibility. Moreover, the nanoprobe could execute logic identification on the cell surface for precise drug delivery. Only if both in the acidic microenvironment and aptamer-targeting marker are present on the cell surface, the sgc8-ASO-chimera strand, carrying an antisense oligonucleotide drug, was released from the nanoprobe and entered into targeted cancer cells for gene silence. Additionally, the in situ drug release facilitated the uptake of drugs mediated by the interaction between sgc8 aptamer and membrane proteins, resulting in enhanced inhibition of cancer cell migration and proliferation. This logic nanoprobe will provide inspiration for designing smart devices for diagnosis of pH-related diseases and targeted drug delivery.


Assuntos
DNA , Sistemas de Liberação de Medicamentos , DNA/química , Sistemas de Liberação de Medicamentos/métodos , Oligonucleotídeos , Membrana Celular , Concentração de Íons de Hidrogênio
6.
Chem Sci ; 14(47): 13860-13869, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-38075660

RESUMO

Accurately quantifying microRNA levels in vivo is of great importance for cancer staging and prognosis. However, the low abundance of microRNAs and interference from the complex tumor microenvironment usually limit the real-time quantification of microRNAs in vivo. Herein, for the first time, we develop an ultrasensitive microRNA (miR)-21 activated ratiometric nanoprobe for quantification of the miR-21 concentration in vivo without signal amplification as well as dynamic tracking of its distribution. The core-satellite nanoprobe by miR-21 triggered in situ self-assembly was built on nanogapped gold nanoparticles (AuNNP probe) and gold nanoparticles (AuNP probe). The AuNP probe generated a photoacoustic (PA) signal and ratiometric SERS signal with the variation of miR-21, whereas the AuNNP probe served as an internal standard, enabling ratiometric SERS imaging of miR-21. The absolute concentration of miR-21 in MCF-7 tumor-bearing mice was quantified to be 83.8 ± 24.6 pM via PA and ratiometric SERS imaging. Our strategy provides a powerful approach for the quantitative detection of microRNAs in vivo, providing a reference for the clinical treatment of cancer.

7.
Biomater Sci ; 11(22): 7423-7431, 2023 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-37815807

RESUMO

Photodynamic therapy (PDT) has the characteristics of being simple and non-invasive, and with on-demand light control. However, most photosensitizers exhibit strong hydrophobicity, low quantum yields in water and low tumor selectivity. In this study, carbon network-hosted porphyrins (CPs) with high biocompatibility and efficient singlet oxygen (1O2) generation were developed to reduce the biotoxicity of photosensitizers and avoid quenching caused by hydrophobic aggregation for enhanced PDT. The CPs were prepared by a simple solid-phase synthesis method using porphyrin, green non-toxic citric acid and urea as the raw materials. The CPs exhibited excellent water solubility and high biocompatibility. Even when the concentration reached 1.5 mg mL-1, cells still had good biological activity. By separately fixing the porphyrins in the carbon network, the CPs avoided aggregation-induced inactivation and had high generation efficiency of 1O2. Furthermore, in order to improve the PDT effect, the CPs were modified with the upper nuclear targeting peptide TAT (T-CPs), which was used to target the nucleus and generate 1O2in situ to directly destroy genetic material. The proposed strategy provides a simple and green path to prepare nanophotosensitizers with high biocompatibility and efficient 1O2 generation for PDT.


Assuntos
Fotoquimioterapia , Porfirinas , Fármacos Fotossensibilizantes/química , Porfirinas/química , Fotoquimioterapia/métodos , Carbono , Água
8.
Nano Lett ; 23(18): 8725-8733, 2023 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-37695255

RESUMO

Inducing pyroptosis in cancer cells holds great potential in cancer immunotherapy. Lipopolysaccharide (LPS)-sensing noncanonical pathways are an important mechanism of pyroptosis to eliminate damaged cells, which has not yet been explored for cancer immunotherapy. Here, we utilize bacterial outer membrane vesicles (OMVs) as a natural LPS carrier to trigger a noncanonical pyroptosis pathway for immunotherapy. To address the concern of systemic toxicity, molecule engineered OMVs were designed by equipping DNA aptamers on the OMVs (Apt-OMVs). In addition to improving capacity to target tumors, Apt-OMVs also took advantage of the spherical nucleic acid structure to shield OMVs against nonspecific immune recognition and evade immunogenicity. The selective pyroptosis enhanced tumor immunogenicity, not only promoting the infiltration of effector T cells but also reducing the amount of immunosuppressive regulatory T cells, which remarkably suppressed tumor growth. This work reports the first pyroptosis inducer by the noncanonical pathway, offering inspiration for safe and efficient pyroptosis-mediated immunotherapy.


Assuntos
Lipopolissacarídeos , Neoplasias , Piroptose , Imunoterapia , Neoplasias/terapia
9.
ACS Nano ; 17(18): 18114-18127, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37695697

RESUMO

In personalized cancer immunotherapy, developing an effective neoantigen nanovaccine with high immunogenicity is a significant challenge. Traditional nanovaccine delivery systems often require nanocarriers, which can hinder the delivery of the neoantigen and cause significant toxicity. In this study, we present an innovative strategy of carrier-free nanovaccine achieved through direct self-assembly of 2'-fluorinated CpG (2'F-CpG) with melanoma neoantigen peptide (Obsl1). Molecular dynamics simulations demonstrated that the introduction of a fluorine atom into CpG increases the noncovalent interaction between 2'F-CpG and Obsl1, which enhanced the loading of Obsl1 on 2'F-CpG, resulting in the spontaneous formation of a hybrid 2'F-CpG/Obsl1 nanovaccine. This nanovaccine without extra nanocarriers showed ultrahigh Obsl1 loading up to 83.19 wt %, increasing the neoantigen peptide uptake by antigen-presenting cells (APCs). In C57BL/6 mice models, we demonstrated the long-term preventive and therapeutic effects of the prepared 2'F-CpG/Obsl1 nanovaccine against B16F10 melanoma. Immunocellular analysis revealed that the nanovaccine activated innate and adaptive immune responses to cancer cells. Hence, this study established a simple, safe, and effective preparation strategy for a carrier-free neoantigen nanovaccine, which could be adapted for the future design of personalized cancer vaccines in clinical settings.


Assuntos
Melanoma , Camundongos , Animais , Camundongos Endogâmicos C57BL , Melanoma/terapia , Células Apresentadoras de Antígenos , Transporte Biológico , Peptídeos
10.
Acta Biomater ; 170: 401-414, 2023 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-37625679

RESUMO

The first approved RNAi therapeutics, ONPATTRO, in 2017 moves the concept of RNA interference (RNAi) therapy from research to clinical reality, raising the hopes for the treatment of currently incurable diseases. However, RNAi therapeutics are still facing two main challenges-susceptibility to enzymatic degradation and low ability to escape from endo/lysosome into the cytoplasm. Therefore, we developed disulfide-based nanospheres (DBNPs) as universal vehicles to achieve efficient RNA delivery to address these problems. Notably, the DBNPs possess unique and desirable features, including improved resistance to nuclease degradation, direct cytoplasmic delivery through thiol-mediated cellular uptake, and cytosolic environment-responsive release, greatly enhancing the bioavailability of RNA therapeutics. Additionally, DBNPs are superior in terms of overcoming formidable physiological barriers, including vascular barriers and impermeable tumor tissues. Owning to these advantages, the DBNPs exhibit efficient gene silencing effect when delivering either small interfering RNA (siRNA) or microRNA in various cell lines and generate remarkable growth inhibition in the zebrafish and mouse model of pancreatic tumors as compared to traditional delivery vectors, such as PEI. Therefore, DBNPs have potential application prospect in RNAi therapy both in vitro and in vivo. STATEMENT OF SIGNIFICANCE: RNA interference (RNAi) therapeutics could target and alter any disease-related mRNA translation, thus have great potential in clinical application. Delivery efficiency of RNA modalities into cell cytoplasm is the main problem that currently limit RNAi therapeutics to release their full potential. Most of the known delivery materials suffer from the endo/lysosomal entrapment and enzymatic degradation during endocytosis-dependent uptake, resulting unsatisfied efficiency of the cytoplasmic release. Here, we developed disulfide-based nanospheres could directly transfer RNA modalities into the cytoplasm and significantly enhance the delivery efficiency, thus holding great potential in RNAi therapy.


Assuntos
Terapêutica com RNAi , Peixe-Zebra , Animais , Camundongos , Interferência de RNA , Terapêutica com RNAi/métodos , RNA Interferente Pequeno/genética , Terapia Genética , Lisossomos , Dissulfetos
11.
Adv Healthc Mater ; 12(28): e2301437, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37379009

RESUMO

Apoptosis has gained increasing attention in cancer therapy as an intrinsic signaling pathway, which leads to minimal leakage of waste products from a dying cell to neighboring normal cells. Among various stimuli to trigger apoptosis, mild hyperthermia is attractive but confronts limitations of non-specific heating and acquired resistance from elevated expression of heat shock proteins. Here, a dual-stimulation activated turn-on T1 imaging-based nanoparticulate system (DAS) is developed for mild photothermia (≈43 °C)-mediated precise apoptotic cancer therapy. In the DAS, a superparamagnetic quencher (ferroferric oxide nanoparticles, Fe3 O4 NPs) and a paramagnetic enhancer (Gd-DOTA complexes) are connected via the N6-methyladenine (m6 A)-caged, Zn2+ -dependent DNAzyme molecular device. The substrate strand of the DNAzyme contains one segment of Gd-DOTA complex-labeled sequence and another one of HSP70 antisense oligonucleotide. When the DAS is taken up by cancer cells, overexpressed fat mass and obesity-associated protein (FTO) specifically demethylates the m6 A group, thereby activating DNAzymes to cleave the substrate strand and simultaneously releasing Gd-DOTA complex-labeled oligonucleotides. The restored T1 signal from the liberated Gd-DOTA complexes lights up the tumor to guide the location and time of deploying 808 nm laser irradiation. Afterward, locally generated mild photothermia works in concert with HSP70 antisense oligonucleotides to promote apoptosis of tumor cells. This highly integrated design provides an alternative strategy for mild hyperthermia-mediated precise apoptotic cancer therapy.


Assuntos
DNA Catalítico , Compostos Heterocíclicos , Nanopartículas , Neoplasias , Compostos Organometálicos , DNA Catalítico/química , Fototerapia , Nanopartículas/química , Oligonucleotídeos , Oligonucleotídeos Antissenso , Linhagem Celular Tumoral , Neoplasias/diagnóstico por imagem , Neoplasias/terapia
12.
Angew Chem Int Ed Engl ; 62(22): e202302255, 2023 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-36959091

RESUMO

Ferrous iron (Fe2+ ) has more potent hydroxyl radical (⋅OH)-generating ability than other Fenton-type metal ions, making Fe-based nanomaterials attractive for chemodynamic therapy (CDT). However, because Fe2+ can be converted by ferritin heavy chain (FHC) to nontoxic ferric form and then sequestered in ferritin, therapeutic outcomes of Fe-mediated CDT agents are still far from satisfactory. Here we report the synthesis of siRNA-embedded Fe0 nanoparticles (Fe0 -siRNA NPs) for self-reinforcing CDT via FHC downregulation. Upon internalization by cancer cells, pH-responsive Fe0 -siRNA NPs are degraded to release Fe2+ and FHC siRNA in acidic endo/lysosomes with the aid of oxygen (O2 ). The accompanied O2 depletion causes an intracellular pH decrease, which further promotes the degradation of Fe0 -siRNA NPs. In addition to initiating chemodynamic process, Fe2+ -catalyzed ⋅OH generation facilitates endo/lysosomal escape of siRNA by disrupting the membranes, enabling FHC downregulation-enhanced CDT.


Assuntos
Nanopartículas , Neoplasias , Humanos , Ferro/metabolismo , Apoferritinas/metabolismo , Apoferritinas/uso terapêutico , RNA Interferente Pequeno/uso terapêutico , Regulação para Baixo , Radical Hidroxila/metabolismo , Nanopartículas/uso terapêutico , Linhagem Celular Tumoral , Neoplasias/tratamento farmacológico , Peróxido de Hidrogênio/metabolismo
13.
Adv Sci (Weinh) ; 10(8): e2202051, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36683237

RESUMO

Surface-enhanced Raman scattering (SERS) is a feasible and ultra-sensitive method for biomedical imaging and disease diagnosis. SERS is widely applied to in vivo imaging due to the development of functional nanoparticles encoded by Raman active molecules (SERS nanoprobes) and improvements in instruments. Herein, the recent developments in SERS active materials and their in vivo imaging and biosensing applications are overviewed. Various SERS substrates that have been successfully used for in vivo imaging are described. Then, the applications of SERS imaging in cancer detection and in vivo intraoperative guidance are summarized. The role of highly sensitive SERS biosensors in guiding the detection and prevention of diseases is discussed in detail. Moreover, its role in the identification and resection of microtumors and as a diagnostic and therapeutic platform is also reviewed. Finally, the progress and challenges associated with SERS active materials, equipment, and clinical translation are described. The present evidence suggests that SERS could be applied in clinical practice in the future.


Assuntos
Técnicas Biossensoriais , Nanopartículas , Nanopartículas/química , Análise Espectral Raman/métodos , Técnicas Biossensoriais/métodos , Imagem Molecular
14.
J Am Chem Soc ; 145(2): 1108-1117, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36622303

RESUMO

Telomerase has long been considered as a biomarker for cancer diagnosis and a therapeutic target for drug discovery. Detecting telomerase activity in vivo could provide more direct information of tumor progression and response to drug treatment, which, however, is hampered by the lack of an effective probe that can generate an output signal without a tissue penetration depth limit. In this study, using the principle of distance-dependent magnetic resonance tuning, we constructed a telomerase-activated magnetic resonance imaging probe (TAMP) by connecting superparamagnetic ferroferric oxide nanoparticles (SPFONs) and paramagnetic Gd-DOTA (Gd(III) 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) complexes via telomerase-responsive DNA motifs. Upon telomerase-catalyzed extension of the primer in TAMP, Gd-DOTA-conjugated oligonucleotides can be liberated from the surface of SPFONs through a DNA strand displacement reaction, restoring the T1 signal of the Gd-DOTA for a direct readout of the telomerase activity. Here we show that, by tracking telomerase activity, this probe provides consistent monitoring of tumor growth kinetics during progression and in response to drug treatment and enables in situ screening of telomerase inhibitors in whole-animal models. This study provides an alternative toolkit for cancer diagnosis, treatment response assessment, and anticancer drug screening.


Assuntos
Telomerase , Animais , Linhagem Celular Tumoral , Telomerase/metabolismo , Cinética , Imageamento por Ressonância Magnética
16.
Adv Drug Deliv Rev ; 194: 114710, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36708774

RESUMO

Plasmonic nanoparticles and their assemblies have been widely used in biosensing, optical imaging, and biomedicine over the past few decades. Especially in the field of radiotherapy, the physicochemical properties of high-Z plasmonic nanomaterials endow them with the ability to sensitize radiotherapy. Compared with single particles, the assembled structure with tunable properties leads to versatile applications in drug delivery and cancer treatment. In this review, we focus on plasmonic nanoparticles and their assemblies for cancer radiotherapy. First, the sensitization mechanism of plasmonic radiosensitizers is briefly introduced. Subsequently, the recent progress in cancer radiotherapy is systematically discussed according to the structure and shape classification. Finally, the current challenges and future perspectives in this field are also discussed in detail.


Assuntos
Nanopartículas Metálicas , Nanopartículas , Nanoestruturas , Neoplasias , Humanos , Nanoestruturas/química , Imagem Óptica , Nanopartículas Metálicas/química , Neoplasias/terapia
17.
Acc Chem Res ; 56(1): 37-51, 2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36533853

RESUMO

X-ray luminescence is an optical phenomenon in which chemical compounds known as scintillators can emit short-wavelength light upon the excitation of X-ray photons. Since X-rays exhibit well-recognized advantages of deep penetration toward tissues and a minimal autofluorescence background in biological samples, X-ray luminescence has been increasingly becoming a promising optical tool for tackling the challenges in the fields of imaging, biosensing, and theragnostics. In recent years, the emergence of nanocrystal scintillators have further expanded the application scenarios of X-ray luminescence, such as high-resolution X-ray imaging, autofluorescence-free detection of biomarkers, and noninvasive phototherapy in deep tissues. Meanwhile, X-ray luminescence holds great promise in breaking the depth dependency of deep-seated lesion treatment and achieving synergistic radiotherapy with phototherapy.In this Account, we provide an overview of recent advances in developing advanced X-ray luminescence for applications in imaging, biosensing, theragnostics, and optogenetics neuromodulation. We first introduce solution-processed lead halide all-inorganic perovskite nanocrystal scintillators that are able to convert X-ray photons to multicolor X-ray luminescence. We have developed a perovskite nanoscintillator-based X-ray detector for high-resolution X-ray imaging of the internal structure of electronic circuits and biological samples. We further advanced the development of flexible X-ray luminescence imaging using solution-processable lanthanide-doped nanoscintillators featuring long-lived X-ray luminescence to image three-dimensional irregularly shaped objects. We also outline the general principles of high-contrast in vivo X-ray luminescence imaging which combines nanoscintillators with functional biomolecules such as aptamers, peptides, and antibodies. High-quality X-ray luminescence nanoprobes were engineered to achieve the high-sensitivity detection of various biomarkers, which enabled the avoidance of interference from the biological matrix autofluorescence and photon scattering. By marrying X-ray luminescence probes with stimuli-responsive materials, multifunctional theragnostic nanosystems were constructed for on-demand synergistic gas radiotherapy with excellent therapeutic effects. By taking advantage of the capability of X-rays to penetrate the skull, we also demonstrated the development of controllable, wireless optogenetic neuromodulation using X-ray luminescence probes while obviating damage from traditional optical fibers. Furthermore, we discussed in detail some challenges and future development of X-ray luminescence in terms of scintillator synthesis and surface modification, mechanism studies, and their other potential applications to provide useful guidance for further advancing the development of X-ray luminescence.


Assuntos
Luminescência , Raios X , Biomarcadores , Diagnóstico por Imagem , Técnicas Biossensoriais , Técnicas de Diagnóstico Molecular
18.
Acta Biomater ; 155: 635-643, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36328129

RESUMO

Non-invasive phototherapy has been emerging as an ambitious tactic for suppression of amyloid-ß (Aß) self-assembly against Alzheimer's disease (AD). However, it remains a daunting challenge to develop efficient photosensitizers for Aß oxygenation that are activatable in a deep brain tissue through the scalp and skull, while reducing side effects on normal tissues. Here, we report an Aß targeted, low-dose X-ray-excitable long-afterglow scintillator (ScNPs@RB/Ab) for efficient deep-brain phototherapy. We demonstrate that the as-synthesized ScNPs@RB/Ab is capable of converting X-rays into visible light to activate the photosensitizers of rose bengal (RB) for Aß oxygenation through the scalp and skull. We show that the ScNPs@RB/Ab persistently emitting visible luminescence can substantially minimize the risk of excessive X-ray exposure dosage. Importantly, peptide KLVFFAED-functionalized ScNPs@RB/Ab shows a blood-brain barrier permeability. In vivo experimental results validated that ScNPs@RB/Ab alleviated Aß burden and slowed cognitive decline in triple-transgenic AD model mice at extremely low X-ray doses without side effects. Our study paves a new pathway to develop high-efficiency transcranial AD phototherapy. STATEMENT OF SIGNIFICANCE: Non-invasive phototherapy has been emerging as an ambitious tactic for suppression of amyloid-ß (Aß) self-assembly against Alzheimer's disease (AD). However, it remains a daunting challenge to develop efficient photosensitizers for Aß oxygenation that are activatable in a deep brain tissue through the scalp and skull, while reducing side effects on normal tissues. Herein, we report an Aß targeted, low-dose X-ray-excitable long-afterglow scintillators (ScNPs@RB/Ab) for efficient deep-brain phototherapy. In vivo experimental results validated that ScNPs@RB/Ab alleviated Aß burden and slowed cognitive decline in triple-transgenic AD model mice at extremely low X-ray doses without side effects.


Assuntos
Doença de Alzheimer , Camundongos , Animais , Doença de Alzheimer/metabolismo , Raios X , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Fototerapia/métodos , Peptídeos beta-Amiloides/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Camundongos Transgênicos , Modelos Animais de Doenças
19.
Chem Sci ; 13(43): 12840-12850, 2022 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-36519050

RESUMO

Radiosensitizers potentiate the radiotherapy effect while effectively reducing the damage to healthy tissues. However, limited sample accumulation efficiency and low radiation energy deposition in the tumor significantly reduce the therapeutic effect. Herein, we developed multifunctional photocatalysis-powered dandelion-like nanomotors composed of amorphous TiO2 components and Au nanorods (∼93 nm in length and ∼16 nm in outer diameter) by a ligand-mediated interface regulation strategy for NIR-II photoacoustic imaging-guided synergistically enhanced cancer radiotherapy. The non-centrosymmetric nanostructure generates stronger local plasmonic near-fields close to the Au-TiO2 interface. Moreover, the Au-TiO2 Schottky heterojunction greatly facilitates the separation of photogenerated electron-hole pairs, enabling hot electron injection, finally leading to highly efficient plasmon-enhanced photocatalytic activity. The nanomotors exhibit superior motility both in vitro and in vivo, propelled by H2 generated via NIR-catalysis on one side of the Au nanorod, which prevents them from returning to circulation and effectively improves the sample accumulation in the tumor. Additionally, a high radiation dose deposition in the form of more hydroxyl radical generation and glutathione depletion is authenticated. Thus, synergistically enhanced radiotherapeutic efficacy is achieved in both a subcutaneous tumor model and an orthotopic model.

20.
Anal Chem ; 94(51): 18009-18016, 2022 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-36519891

RESUMO

Traditional spherical nucleic acids (SNAs) based on gold nanoparticles (AuNPs) assembled through Au-S covalent bonds are widely used in DNA-programmable assembly, biosensing, imaging, and therapeutics. However, biological thiols and other chemical substances can break the Au-S bonds and cause response distortion during the application process, specifically in cell environments. Herein, we report a new type of SNAs based on 2'-fluorinated DNA-functionalized AuNPs with excellent colloidal stability under high salt conditions (up to 1 M NaCl) and over a broad pH range (1-14), as well as resistance to biothiols. The fluorinated spherical nucleic acid probe (Au/FDNA probe) could detect targeted cancer cells with high fidelity. Compared to the traditional thiolated DNA-functionalized AuNP probe (Au-SDNA probe), the Au/FDNA probe exhibited a higher sensitivity to the target and a lower signal-to-background ratio. Furthermore, the Au/FDNA probe could discriminate target cancer cells in a mixed culture system. Using the proposed FDNA functionalization method, previously developed SNAs based on AuNPs could be directly adapted, which might open a new avenue for the design and application of SNAs.


Assuntos
Técnicas Biossensoriais , Nanopartículas Metálicas , Ácidos Nucleicos , Ouro/química , Nanopartículas Metálicas/química , DNA/química , Ácidos Nucleicos/química , Sondas de DNA/química , Técnicas Biossensoriais/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA