Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Nano Lett ; 23(8): 3444-3450, 2023 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-37014732

RESUMEN

Highly doped lanthanide luminescent nanoparticles exhibit unique optical properties, providing exciting opportunities for many ground-breaking applications, such as super-resolution microscopy, deep-tissue bioimaging, confidentiality, and anticounterfeiting. However, the concentration-quenching effect compromises their luminescence efficiency/brightness, hindering their wide range of applications. Herein, we developed a low-temperature suppression cross-relaxation strategy, which drastically enhanced upconversion luminescence (up to 2150-fold of green emission) in Er3+-rich nanosystems. The cryogenic field opens the energy transport channel of Er3+ multiphoton upconversion by further suppressing phonon-assisted cross-relaxation. Our results provide direct evidence for understanding the energy loss mechanism of photon upconversion, deepening a fundamental understanding of the upconversion process in highly doped nanosystems. Furthermore, it also suggests the potential applications of upconversion nanoparticles for extreme ambient-temperature detection and anticounterfeiting.

2.
Nano Lett ; 23(13): 6241-6248, 2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37098101

RESUMEN

Smart control of ionic interactions is a key factor to manipulate the luminescence dynamics of lanthanides and tune their emission colors. However, it remains challenging to gain a deep insight into the physics involving the interactions between heavily doped lanthanide ions and in particular between the lanthanide sublattices for luminescent materials. Here we report a conceptual model to selectively manipulate the spatial interactions between erbium and ytterbium sublattices by designing a multilayer core-shell nanostructure. The interfacial cross-relaxation is found to be a leading process to quench the green emission of Er3+, and red-to-green color-switchable upconversion is realized by fine manipulation of the interfacial energy transfer on the nanoscale. Moreover, the temporal control of up-transition dynamics can also lead to an observation of green emission due to its fast rise time. Our results demonstrate a new strategy to achieve orthogonal upconversion, showing great promise in frontier photonic applications.

3.
Nano Lett ; 22(13): 5339-5347, 2022 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-35708527

RESUMEN

Combating the concentration quenching effect by increasing the concentration of sensitized rare-earth ions in rational design upconversion nanostructure will make it easier to utilize injection energy flux and transfer it to emitters, resulting in improved upconversion luminescence (UCL). We proposed a host-sensitized nanostructure (active core@luminescent shell@inert shell) to improve multiphoton UCL of Tm3+ based on the LiLnF4 host. Yb3+ ions were isolated in the core as energy absorbents, and Tm3+ was doped in the interior LiYbF4 host shell. Compared with sandwich structured nanocrystals (Y@Y:Yb/Tm@Y), reverse structure (YbTm@Yb@Y), and fully doped structure (YbTm@YbTm@Y), the proposed structure achieved the highest efficiency of multiphoton UCL and favored a better FRET-based application performance as the Tm3+ located at an optimized spatial distribution. Furthermore, steady-state and dynamic analysis results demonstrate that by manipulating the spatial distribution of the active ions, excited energy can be tuned to enable multiphoton upconversion enhancement, overcoming the conventional limitations.


Asunto(s)
Nanopartículas , Nanoestructuras , Luminiscencia , Nanopartículas/química
4.
RSC Adv ; 14(28): 20152-20162, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38915327

RESUMEN

Synergistic photodynamic therapy (PDT) with other therapeutic modalities can enhance the therapeutic efficacy of tumor treatment and reduce the adverse effects associated with drug leakage and off-target accumulation. However, shaping combined strategies for synergistic therapy remains challenging. Herein, we developed versatile hybrid liposomes self-assembled from Ce6-lipid conjugates and loaded with the chemo drug doxorubicin (DOX) and ferroptosis inducer Fe3O4 nanoparticles for synergistic PDT/chemo/ferroptosis therapy. Abundant ROS are generated by PDT upon 650 nm light irradiation, Fe3O4-mediated Fenton reaction, and DOX-induced apoptosis. Furthermore, amplifying oxidative stress in cancer cells to disrupt cellular redox homeostasis could accelerate tumor cell death through oxidative damage to lipids, proteins, and DNA. Overall, this work highlights liposome-based therapeutic nanoformulations, thus offering a breakthrough redox homeostasis-based synergistic PDT/chemo/ferroptosis therapy for lung cancer.

5.
Histol Histopathol ; 38(9): 1055-1068, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36546683

RESUMEN

BACKGROUND: Cisplatin is the first-line chemotherapy drug for the treatment of gastric cancer (GC) patients. However, GC patients who are resistant to cisplatin often do not benefit from it. Therefore, finding a key molecule that affects cisplatin sensitivity is expected to enhance the efficacy of cisplatin in GC treatment. METHODS: The human GC cell lines SGC-7901 and BGC-823 were used. The protein chip array was used to screen the cisplatin-resistance genes from the complete response and non-complete response GC patients' tissues, then, the differential gene expression analysis, GO function annotation analysis, and KEGG pathway enrichment analysis were performed. The GC tissue chip in the GEO database was analyzed to screen the target gene. Flow cytometry, Hoechst 33342 staining assay, Western Blot, MTT, tumor sphere formation, cell cycle, and apoptosis assays were performed to explore the effect of Phospholysine Phosphohistidine Inorganic Pyrophosphate Phosphatase (LHPP) on the apoptosis, stemness, and reactive oxygen species (ROS) accumulation of cisplatin-resistant GC cells treated with cisplatin. In vivo, the cisplatin-resistant GC cell lines transfected with pcDNA-LHPP or si-LHPP were injected subcutaneously into mice to construct GC subcutaneous xenograft GC models. RESULTS: Based on protein chip array and bioinformatics analysis, it was found that LHPP is the core molecule in the cisplatin resistance regulatory network in GC, and its expression is down-regulated in GC cisplatin-resistant tissues and cells. In vitro and in vivo experimental results show that the up-regulated expression of LHPP is closely related to the increase in sensitivity of GC to cisplatin. Mechanically, we found that overexpression of LHPP may inhibit the activation of the JNK and p38 MAPK pathways, promote cisplatin-induced ROS accumulation, suppress stemness, and enhance the sensitivity of GC to cisplatin. CONCLUSIONS: Up-regulation of LHPP may inhibit the activation of the JNK and p38 MAPK pathways, attenuate stemness, and enhance the accumulation of intracellular ROS, thereby promoting cisplatin-mediated GC cell apoptosis and enhancing cisplatin sensitivity.


Asunto(s)
Cisplatino , Neoplasias Gástricas , Animales , Humanos , Ratones , Apoptosis , Línea Celular Tumoral , Proliferación Celular/genética , Cisplatino/farmacología , Regulación Neoplásica de la Expresión Génica , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Neoplasias Gástricas/genética , Pirofosfatasa Inorgánica/metabolismo
6.
BMC Med Genomics ; 16(1): 93, 2023 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-37138287

RESUMEN

BACKGROUND: The interaction between tumor cells and immune or non-immune stromal cells creates a unique tumor microenvironment, which plays an important role in the growth, invasion and metastasis of gastric cancer (GC). METHODS: The candidate genes were selected to construct risk-score by univariate and multivariate Cox regression analysis. Nomograms were constructed by combining clinical pathological factors, and the model performance was evaluated by receiver operating characteristic curve, decision curve analysis, net reclassification improvement and integrated discrimination improvement. The functional enrichment between high-risk group (HRisk) and low-risk group (LRisk) was explored through GO, KEGG, GSVA and ssGSEA. CIBERSORT, quanTIseq and xCell were used to explore the immune cell infiltration between HRisk and LRisk. The relevant EMT scores, macrophage infiltration scores and various metabolic scores were calculated through the "IOBR" package and analyzed visually. RESULTS: Through univariate and multivariate Cox regression analysis, we obtained the risk-score of fittings six lipid metabolism related genes (LMAGs). Through survival analysis, we found that risk-score has significant prognostic significance and can accurately reflect the metabolic level of patients. The AUCs of the nomogram model incorporating risk-score 1, 3 and 5 years were 0.725, 0.729 and 0.749 respectively. In addition, it was found that the inclusion of risk-score could significantly improve the prediction performance of the model. It was found that the arachidonic acid metabolism and prostaglandin synthesis were up-regulated in HRisk, and more tumor metastasis related markers and immune related pathways were also enriched. Further study found that HRisk had higher immune score and M2 macrophage infiltration. More importantly, the immune checkpoints of tumor associated macrophages involved in tumor antigen recognition disorders increased significantly. We also found that ST6GALNAC3 can promote arachidonic acid metabolism and up-regulate prostaglandin synthesis, increase M2 macrophage infiltration, induce epithelial mesenchymal transformation, and affect the prognosis of patients. CONCLUSIONS: Our research found a novel and powerful LMAGs signature. Six-LMAGs features can effectively evaluate the prognosis of GC patients and reflect the metabolic and immune status. ST6GALNAC3 may be a potential prognostic marker to improve the survival rate and prognostic accuracy of GC patients, and may even be a potential biomarker of GC patients, indicating the response to immunotherapy.


Asunto(s)
Metabolismo de los Lípidos , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , Pronóstico , Ácido Araquidónico , Biomarcadores de Tumor/genética , Prostaglandinas , Microambiente Tumoral
7.
Acta Biomater ; 171: 519-531, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37714248

RESUMEN

Periodontitis is a biofilm-related disease characterized by damage to the periodontal tissue and the development of systemic diseases. However, treatment of periodontitis remains unsatisfactory, especially with deep-tissue infections. This study describes rationally designed multifunctional photothermocatalytic agents for near-infrared-II light-mediated synergistic antibiofilm treatment, through modification of Lu-Bi2Te3 with Fe3O4 and poly(ethylene glycol)-b-poly(l-arginine) (PEG-b-PArg). Notably, 1064-nm laser irradiation led to photothermal/thermocatalytic effects, resulting in the synergistic generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) and consequent damage to the biofilm. This treatment was based on the thermoelectric and photothermal conversion properties of Lu-Bi2Te3, the peroxidase-like catalytic capacity of Fe3O4, and the guanidinium polymer, PEG-b-PArg. Oxidative damage to biofilm was further enhanced by H2O2, resulting in the effective elimination of biofilm both in vitro and in vivo. These findings suggest that this synergistic therapeutic strategy is effective for the clinical treatment of periodontitis. STATEMENT OF SIGNIFICANCE: The current treatment for periodontitis involves time-consuming and labor-intensive clinical scaling of the teeth. The present study is the first to assess the efficacy of a photothermal catalyst for periodontitis treatment. This used near-infrared-II light at 1064 nm to induce oxidative damage in the biofilm, resulting in its degradation. The synergistic photothermal/thermoelectric effect produced deep tissue penetration and was well tolerated, and can kill the biofilm formed by periodontitis pathogens up to 5 orders of magnitude, effectively treating the biofilm-induced periodontitis.


Asunto(s)
Hipertermia Inducida , Periodontitis , Humanos , Peróxido de Hidrógeno , Periodontitis/terapia , Fototerapia , Estrés Oxidativo
8.
Nat Commun ; 14(1): 1079, 2023 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-36841808

RESUMEN

Fluorescence bioimaging based on rare-earth-doped nanocrystals (RENCs) in the shortwave infrared (SWIR, 1000-3000 nm) region has aroused intense interest due to deeper penetration depth and clarity. However, their downshifting emission rarely shows sufficient brightness beyond 1600 nm, especially in NIR-IIc. Here, we present a class of thulium (Tm) self-sensitized RENC fluorescence probes that exhibit bright downshifting luminescence at 1600-2100 nm (NIR-IIb/c) for in vivo bioimaging. An inert shell coating minimizes surface quenching and combines strong cross-relaxation, allowing LiTmF4@LiYF4 NPs to emit these intense downshifting emissions by absorbing NIR photons at 800 nm (large Stokes shift ~1000 nm with a absolute quantum yield of ~14.16%) or 1208 nm (NIR-IIin and NIR-IIout). Furthermore, doping with Er3+ for energy trapping achieves four-wavelength NIR irradiation and bright NIR-IIb/c emission. Our results show that Tm-based NPs, as NIR-IIb/c nanoprobes with high signal-to-background ratio and clarity, open new opportunities for future applications and translation into diverse fields.


Asunto(s)
Metales de Tierras Raras , Nanopartículas , Luminiscencia , Tulio , Metales de Tierras Raras/química , Nanopartículas/química , Fotones
9.
RSC Adv ; 13(13): 8535-8539, 2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36926301

RESUMEN

Lanthanide-doped upconversion nanoparticles (UCNPs) have attracted great attention in temperature sensing because of their widespread thermal quenching effect (TQE), a phenomenon in which luminescence intensity decreases as the temperature increases. However, enhancing the TQE of activated ions without changing the dopants or the host is still challenging. Herein, Yb3+ and Er3+ codoped UCNPs in a cubic CaGdF5 host were synthesized by a coprecipitation method for optical temperature sensing. Compared with the homogeneous shell (CaGdF5), those heterogeneous (CaF2) shelled UCNPs exhibited stronger upconversion luminescence (UCL) due to the significantly reduced multiphonon nonradiative relaxation. Further, we investigated the effects of homogeneous and heterogeneous shells on TQE. The relationship between the intensity ratio of the green emission bands of Er3+ ions (2H11/2 → 4I15/2 and 4S3/2 → 4I15/2) and temperature are obtained for these two core@shell UCNPs. The results demonstrated that the UCNPs with CaF2 shells are more sensitive to temperature in the 200-300 K. The maximum thermal sensitivity of CaGdF5:Yb,Er@CaF2 could reach 2.2% K-1 at 200 K. These results indicate that the heterogeneous core@shell UCNPs are promising for use as optical temperature sensors.

10.
Adv Healthc Mater ; 11(19): e2200809, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35848849

RESUMEN

Nanozymes with multienzyme-mimicking activities have shown great potential in cancer therapy due to their ability to modulate the complex tumor microenvironment (TME). Herein, a second near-infrared (NIR-II) photothermal-nanocatalyst by decorating Bi2 Te3 nanosheets with ultrasmall Au/Pd bimetallic nanoparticles (Bi2 Te3 -Au/Pd) to reverse the immunosuppressive TME is developed. The peroxidase (POD)-like and catalase (CAT)-like activities, and glutathione (GSH) consumption capacity of Au/Pd modulates the TME by disrupting the intracellular redox homeostasis and relieving hypoxia in the TME. Notably, the amplified oxidative stress induces the accumulation of lipid hydroperoxides (LPO) for enhanced ferroptosis. Moreover, upon NIR-II photoirradiation at 1064 nm, the localized heat generated by Bi2 Te3 not only directly ablates the cancer cells but also enhances the Au/Pd-mediated catalysis-mediated cancer therapy. Furthermore, both in vitro and in vivo studies confirm that the Bi2 Te3 -Au/Pd nanocatalysts (BAP NCs) can effectively suppress tumor growth by inducing immunogenic cell death (ICD), and suppressing metastasis and recurrence by the synergistic treatment. Overall, this study provides a promising theranostic strategy for effective tumor inhibition.


Asunto(s)
Nanopartículas , Neoplasias , Catalasa , Línea Celular Tumoral , Glutatión , Humanos , Lípidos , Neoplasias/tratamiento farmacológico , Medicina de Precisión , Nanomedicina Teranóstica , Microambiente Tumoral
11.
Biomater Sci ; 10(4): 1104-1112, 2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-35044388

RESUMEN

Ferroptosis therapy (FT) is an attractive strategy to selectively damage cancer cells through lipid peroxide (LPO) over-accumulation. However, this therapy suffers from poor therapeutic efficacy due to the limited Fenton reaction efficiency and the evolved intrinsic resistance mechanism in the tumor microenvironment (TME). The exploitation of novel ferroptosis inducers is of significance for improving the efficacy of FT. Here, we develop a plate-like Bi2Se3-Fe3O4/Au (BFA) theranostic nanoplatform, which can increase the Fenton reaction rate to enhance FT in an active-passive way. In detail, benefiting from the internal synergistic effect of Fe3O4 NPs and Au NPs and external NIR-mediated hyperthermia, the BFA NPs can boost hydroxyl radical (˙OH) generation to enhance intracellular oxidative stress and further induce ferroptosis by inactivating glutathione peroxidase 4 (GPX4). Furthermore, the BFA NPs show high photothermal conversion efficiency in both the NIR-I and NIR-II windows (66.2% at 808 nm and 58.2% at 1064 nm, respectively); therefore, as a photothermal agent (PTA), they can also ablate cancer cells directly by NIR-triggered photothermal therapy (PTT). Meanwhile, BFA NPs could be used as an efficient diagnostic agent for photoacoustic (PA)/magnetic resonance (MR)/X-ray imaging to guide the synergistic therapy of photothermal-ferroptosis. Therefore, BFA NP-mediated enhanced photothermal-ferroptosis therapy represents a promising strategy for the application of nanomaterials in tumor therapy.


Asunto(s)
Ferroptosis , Hipertermia Inducida , Nanopartículas , Neoplasias , Línea Celular Tumoral , Humanos , Neoplasias/terapia , Terapia Fototérmica , Nanomedicina Teranóstica , Microambiente Tumoral
12.
J Mater Chem C Mater ; 10(2): 688-695, 2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-35127099

RESUMEN

Collaborative therapy is regarded as an effective approach in increasing the therapeutic efficacy of cancer. In this work, we have proposed and validated the concept of upconversion lumienscence image guided synergy of photodynamic therapy (PDT) and radiotherapy (RT) for deep cancer, via a specially designed nanoplatform integrating near infrared (NIR) light activated luminescence upconversion and X-ray induced scintillation. Upon NIR light irradiation, the nanoplatform emits highly monochromatic red light solely for imaging the targeted cancer cells without triggering therapy; however, when the irradiation turns to a low dose of X-rays, scintillation will occur which induces effectively the PDT destroying the cancer cells together with X-ray induced RT. The novel theranostic nanoplatform is constructed in such a way that the interactions between the upconversion core and the outmost scintillating shell are blocked effectively by an inert layer between them. This structural design not only enables a nearly perfect excitation energy delivery (∼100% at a spectral overlapping wavelength of ∼540 nm) from the outermost scintellating layer to the surface-anchored photosensitizers and so a maximum yield of radical oxygen species, but also achieves a strong NIR induced upconversion luminescence for imaging. Since PDT and RT attack different parts of a cancer cell, this synergy is more effective in destroying cancer than a single therapy, resulting in the reduction of the X-ray irradiation dosage. As a proof of principle, the theranostic effect is validated by in vitro and in vivo experiments, exhibiting the great potential of this sort of nanoplatform in deep cancer treatment.

13.
Light Sci Appl ; 10(1): 105, 2021 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-34011931

RESUMEN

Internal hydroxyl impurity is known as one of the main detrimental factors affecting the upconversion (UC) efficiency of upconversion luminescence (UCL) nanomaterials. Different from surface/ligand-related emission quenching which can be effectively diminished by, e.g., core/shell structure, internal hydroxyl is easy to be introduced in synthesis but difficult to be quantified and controlled. Therefore, it becomes an obstacle to fully understand the relevant UC mechanism and improve UC efficiency of nanomaterials. Here we report a progress in quantifying and large-range adjustment of the internal hydroxyl impurity in NaYF4 nanocrystals. By combining the spectroscopy study and model simulation, we have quantitatively unraveled the microscopic interactions underlying UCL quenching between internal hydroxyl and the sensitizers and activators, respectively. Furthermore, the internal hydroxyl-involved UC dynamical process is interpreted with a vivid concept of "Survivor effect," i.e., the shorter the migration path of an excited state, the larger the possibility of its surviving from hydroxyl-induced quenching. Apart from the consistent experimental results, this concept can be further evidenced by Monte Carlo simulation, which monitors the variation of energy migration step distribution before and after the hydroxyl introduction. The new quantitative insights shall promote the construction of highly efficient UC materials.

14.
ACS Nano ; 15(12): 20643-20655, 2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34878760

RESUMEN

Photodynamic therapy (PDT) has been widely used in tumor therapy due to its high spatial-temporal control and noninvasiveness. However, its clinical application is limited by weak efficacy, shallow tissue penetration, and phototoxicity. Herein, a facile theranostic nanoplatform based on photoswitchable lanthanide-doped nanoparticles was designed. Typically, these nanoparticles had UV-blue and 1525 nm emission upon 980 nm excitation and 1525 nm emission upon 800 nm excitation. We further used these nanoparticles for achieving real-time near-infrared (NIR)-IIb imaging (800 nm) with a high signal-to-noise ratio and imaging-guided PDT (980 nm). Moreover, such a photoswitchable nanoplatform capping with pH-sensitive calcium phosphate for coloading doxorubicin (a chemotherapeutic immunogenic cell death [ICD] inducer) and paramagnetic Mn2+ ions enhances T1-magnetic resonance imaging in the tumor microenvironment. Our results suggest that this theranostic nanoplatform could not only kill tumor cells directly through dual-modal image-guided PDT/chemotherapy but also inhibit distant tumor and lung metastasis through ICD. Therefore, it has great potential for clinical application .


Asunto(s)
Nanopartículas , Neoplasias , Fotoquimioterapia , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Humanos , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Nanomedicina Teranóstica , Microambiente Tumoral
15.
J Mater Chem B ; 7(4): 548-555, 2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-32254788

RESUMEN

Synergistic thermo-chemotherapy based multiple stimuli-responsive drug delivery systems have achieved significant improvement of cancer curative effects compared with single modality treatment. Nevertheless, the efficacy of thermo-chemotherapy is often reduced in drug-resistant tumors and the therapy method is unexpectedly associated with potential toxicity by utilizing poorly degradable materials. Here, we report a simple approach to encapsulate three drug payloads into multi-sensitive and degradable nanospheres (SDC@NS) to achieve anticancer effects. SDC@NS comprise a photothermal agent (cypate), an anticancer agent (doxorubicin), and a nitric oxide donor (SNAP) to achieve controllable drugs release in high concentration glutathione or under near-infrared light (NIR) irradiation. Hyperthermia from NIR-mediated cypate can accelerate cancer cell apoptosis in vitro and tumor tissue ablation in vivo. Furthermore, our results also confirmed that the nitric oxide-based SDC@NS showed significant cytotoxicity compared to the nitric oxide absent group (denoted as DC@NS) and an enhanced chemotherapy effect in vivo. The photothermal effect and payloads can synchronously realize cancer therapy and provide a new insight into the enhanced synergistic therapeutic effect.


Asunto(s)
Terapia Combinada/métodos , Sistemas de Liberación de Medicamentos/métodos , Rayos Infrarrojos/uso terapéutico , Nanosferas/uso terapéutico , Neoplasias/terapia , Animales , Apoptosis/efectos de los fármacos , Doxorrubicina/administración & dosificación , Liberación de Fármacos , Humanos , Hipertermia Inducida , Indoles/farmacología , Células LLC-PK1 , Células MCF-7 , Ratones Endogámicos C57BL , Óxido Nítrico/farmacología , Propionatos/farmacología , Porcinos , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Chin J Nat Med ; 16(2): 81-89, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29455732

RESUMEN

Andrographis paniculata (Burm. f.) Nees (AP) is commonly used for the treatment of many infectious diseases and has been cultivated widely in Asian countries, and has been included in United States Pharmacopoeia as a dietary supplement, but the cultivars of A. paniculata are not abundant due to its self-pollinated. With the aims to enrich AP resources and provide materials for after breeding we explored the polyploidy induction. Different explants, colchicine concentration, and treatment time were tested. After identification by flow cytometry, eleven polyploid plants with different morphologic traits were obtained. The agronomic traits and andrographolide concentration of the polyploids were improved greatly. One of the polyploids (serial 3-7) was chosen for further study. The traits of the second and third generation polyploids (serial 3-7) were stable. Compared with the normal plants, the seeds (2nd generation) weight increased by 31%, and the andrographolide concentration of the leaves increased by 14% (2nd) and 28% (3rd). In conclusion, AP autopolyploids with different morphologic traits were established successfully for the first time, and the polyploids induction might be effective for crop improvement of AP.


Asunto(s)
Andrographis/crecimiento & desarrollo , Andrographis/genética , Extractos Vegetales/química , Poliploidía , Andrographis/química , Cruzamiento , Técnicas de Cultivo de Célula
18.
ACS Nano ; 12(4): 3217-3225, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29489327

RESUMEN

Photoswitchable materials are important in broad applications. Recently appeared inorganic photoswitchable upconversion nanoparticles (PUCNPs) become a competitive candidate to surmount the widespread issue of the organic counterparts -photobleaching. However, current PUCNPs follow solely Yb3+/Nd3+ cosensitizing mode, which results in complex multilayer doping patterns and imperfectness of switching in UV-blue region. In this work, we have adopted a new strategy to construct Nd3+ free PUCNPs-NaErF4@NaYF4@NaYbF4:0.5%Tm@NaYF4. These PUCNPs demonstrate the superior property of photoswitching. A prominent UV-blue emission from Tm3+ is turned on upon 980 nm excitation, which can be completely turned off by 800 nm light. The quasi-monochromatic red upconversion emission upon 800 nm excitation-a distinct feature of undoping NaErF4 upconversion system-endows the PUCNPs with promising image-guided photoinduced "off-on" therapy in biomedicine. As a proof-of-concept we have demonstrated the imaging-guided photodynamic therapy (PDT) of cancer, where 800 nm excitation turns off the UV-blue emission and leaves the emission at 660 nm for imaging. Once the tumor site is targeted, excitation switching to 980 nm results in UV-blue emission and the red emission. The former is used to induce PDT, whereas the latter is to monitor the therapeutic process. Our study implies that this upconversion photoswitching material is suitable for real-time imaging and image-guided therapy under temporal and spatial control.


Asunto(s)
Nanopartículas/química , Fármacos Fotosensibilizantes/química , Rayos Infrarrojos , Fotoquimioterapia , Rayos Ultravioleta
19.
Nanoscale ; 10(26): 12356-12363, 2018 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-29694473

RESUMEN

Multimodality imaging-guided therapy based on lanthanide-doped upconversion nanoparticles (UCNPs) has become a trend in cancer theranostics. However, the overheating effect of 980 nm excitation in photodynamic therapy (PDT) and the difficulties in optimizing multimodality imaging integration within a single particle are still challenges. Herein, 800 nm driven NaErF4@NaLuF4 UCNPs have been explored for optimized multimodality imaging and near-infrared (NIR) triggered PDT. Our results confirmed that the optimal ∼5 nm shell thickness can well balance the enhancement of upconversion luminescence and the attenuation of energy transfer efficiency from Er3+ towards a photosensitizer, to achieve efficient production of singlet oxygen (1O2) for PDT under 800 nm excitation. Furthermore, the as-obtained NaErF4@NaLuF4 UCNPs showed effective and applicable performance for upconversion luminescence (UCL) imaging, X-ray computed tomography (CT), and high-field T2 magnetic resonance imaging (MRI). This nanomaterial can serve as an excellent theranostic agent for multimodality imaging and image-guided therapy.


Asunto(s)
Elementos de la Serie de los Lantanoides , Imagen Multimodal , Nanopartículas , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/tratamiento farmacológico , Fotoquimioterapia , Células A549 , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Fármacos Fotosensibilizantes
20.
Nanoscale ; 9(23): 7941-7946, 2017 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-28569319

RESUMEN

It is generally accepted that a lanthanide ions based upconversion material follows an activator low doping strategy (normally <3 mol%), because of the restriction of the harmful concentration quenching effect. Here, we demonstrate that this limitation can be broken in nanostructures. Simply by using an inert shell coating strategy, the concentration quenching effect for the activator (Er3+) could be eliminated and highly efficient upconversion luminescence realized in the activator fully doped nanostructure, e.g. NaErF4@NaYF4. More importantly, this novel nanostructure achieves some long-cherished desires, such as multiple-band co-excitation (∼800 nm, ∼980 nm and ∼1530 nm) and monochromic red emission. Proof-of-concept experiments are presented of the potential benefit of this structure in solar cells and anti-counterfeiting. This nanostructure offers new possibilities in realizing high upconversion emission and novel functionalities of lanthanide based nanomaterials.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA