Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 218
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell Neurosci ; : 103957, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39111720

RESUMEN

BACKGROUND: Microglia is the primary source of inflammatory factors during migraine attacks. This study aims to investigate the role of microglia related genes (MRGs) in migraine attacks. METHODS: The RNA sequencing results of migraineurs and the panglaodb database were used to obtain differentially expressed genes (DEGs) in migraine related to microglia. A migraine rat model was established for validating and localizing of the MRGs, and subsequent screening for target genes was conducted. A shRNA was designed to interference the expression of target genes and administered into the trigeminal ganglion (TG) of rats. Pain sensitivity in rats was evaluated via the hot water tail-flick (HWTF) and formalin-induced pain (FIP) experiments. ELISA was used to quantify the levels of inflammatory cytokines and CGRP. WB and immunofluorescence assays were applied to detect the activation of microglia. RESULTS: A total of five DEGs in migraine related to microglia were obtained from RNA sequencing and panglaodb database. Animal experiments showed that these genes expression were heightened in the TG and medulla oblongata (MO) of migraine rats. The gene S100A8 co-localized with microglia in both TG and MO. The HWTF and FIP experiments demonstrated that interference with S100A8 alleviated the sense of pain in migraine rats. Moreover, the levels of TNFα, IL-1ß, IL-6, and CGRP in the TG and MO of rats in the model rats were increased, and the expression of microglia markers IBA-1, M1 polarization markers CD86 and iNOS was upregulated. Significantly, interference with S100A8 reversed these indicators. CONCLUSION: Interference with S100A8 in microglia increased the pain threshold during migraine attacks, and inhibited neuroinflammation and microglia activation.

2.
Proc Natl Acad Sci U S A ; 119(10): e2107453119, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35239437

RESUMEN

SignificanceEpidermal growth factor receptor (EGFR) is one of the most important membrane receptors that transduce growth signals into cells to sustain cell growth, proliferation, and survival. EGFR signal termination is initiated by EGFR internalization, followed by trafficking through endosomes, and degradation in lysosomes. How this process is regulated is still poorly understood. Here, we show that hepatocyte growth factor regulated tyrosine kinase substrate (HGS), a key protein in the EGFR trafficking pathway, is dynamically modified by a single sugar N-acetylglucosamine. This modification inhibits EGFR trafficking from endosomes to lysosomes, leading to the accumulation of EGFR and prolonged signaling. This study provides an important insight into diseases with aberrant growth factor signaling, such as cancer, obesity, and diabetes.


Asunto(s)
Endosomas/metabolismo , Lisosomas/metabolismo , Transducción de Señal , Acilación/genética , Endosomas/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Células Hep G2 , Humanos , Lisosomas/genética , Transporte de Proteínas/genética
3.
Small ; 20(8): e2306378, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37817359

RESUMEN

Proteolysis-targeting chimeras (PROTACs) can provide promising opportunities for cancer treatment, while precise regulation of their activities remains challenging to achieve effective and safe therapeutic outcomes. A semiconducting polymer nanoPROTAC (SPNFeP ) is reported that can achieve ultrasound (US) and tumor microenvironment dual-programmable PROTAC activity for deep-tissue sonodynamic-ferroptosis activatable immunotherapy. SPNFeP is formed through a nano-precipitation of a sonodynamic semiconducting polymer, a ferroptosis inducer, and a newly synthesized PROTAC molecule. The semiconducting polymers work as sonosensitizers to produce singlet oxygen (1 O2 ) via sonodynamic effect under US irradiation, and ferroptosis inducers react with intratumoral hydrogen peroxide (H2 O2 ) to generate hydroxyl radical (·OH). Such a dual-programmable reactive oxygen species (ROS) generation not only triggers ferroptosis and immunogenic cell death (ICD), but also induces on-demand activatable delivery of PROTAC molecules into tumor sites. The effectively activated nanoPROTACs degrade nicotinamide phosphoribosyl transferase (NAMPT) to suppress tumor infiltration of myeloid-derived suppressive cells (MDSCs), thus promoting antitumor immunity. In such a way, SPNFeP mediates sonodynamic-ferroptosis activatable immunotherapy for entirely inhibiting tumor growths in both subcutaneous and 2-cm tissue-covered deep tumor mouse models. This study presents a dual-programmable activatable strategy based on PROTACs for effective and precise cancer combinational therapy.


Asunto(s)
Ferroptosis , Neoplasias , Animales , Ratones , Inmunoterapia , Terapia Combinada , Neoplasias/terapia , Polímeros , Línea Celular Tumoral , Microambiente Tumoral
4.
Langmuir ; 40(32): 17141-17150, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39096500

RESUMEN

In this work, a nonequilibrium molecular dynamics simulation is utilized to explore the effect of network structure of graphene (GE) on the thermal conductivity of the GE/polydimethylsiloxane (PDMS) composite. First, the thermal conductivity of composites rises with increasing volume fraction of GE. The heat transfer ability via the GE channel is found to be nearly the same by analyzing the GE-GE interfacial thermal resistance (ITR). More heat energy is transferred via the GE channel at the high volume fraction of GE by calculating the GE heat transfer ratio, which leads to the high thermal conductivity. Then, the thermal conductivity of composites rises with increasing stacking area between GE, which is attributed to both the strong heat transfer ability via the GE channel and the high GE heat transfer ratio. Following it, the thermal conductivity of composites first rises and then drops down with increasing defect density for a single vacancy defect while it continuously increases for a single void defect. The heat transfer ability between GE is enhanced due to the formation of interlayer covalent bonds. However, the intrinsic thermal conductivity of GE is significantly reduced for a single vacancy defect while it remains relatively well for a single void defect. As a result, the GE heat transfer ratio is maximum at the intermediate defect density for a single vacancy defect while it rises monotonically for a single void defect, which can rationalize the thermal conductivity. Meanwhile, the relationship between ITR and the number of covalent bonds can be described by an empirical equation. Finally, the thermal conductivity for the stacked structure is larger than that for the noncontact structure or the intersected structure. In summary, this work provides a clear and novel understanding of how the network structure of GE influences the thermal conductivity of the GE/PDMS composite.

5.
J Nanobiotechnology ; 22(1): 313, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38840120

RESUMEN

Adoptive cellular immunotherapy as a promising and alternative cancer therapy platform is critical for future clinical applications. Natural killer (NK) cells have attracted attention as an important type of innate immune regulatory cells that can rapidly kill multiple adjacent cancer cells. However, these cells are significantly less effective in treating solid tumors than in treating hematological tumors. Herein, we report the synthesis of a Fe3O4-PEG-CD56/Avastin@Ce6 nanoprobe labeled with NK-92 cells that can be used for adoptive cellular immunotherapy, photodynamic therapy and dual-modality imaging-based in vivo fate tracking. The labeled NK-92 cells specifically target the tumor cells, which increases the amount of cancer cell apoptosis in vitro. Furthermore, the in vivo results indicate that the labeled NK-92 cells can be used for tumor magnetic resonance imaging and fluorescence imaging, adoptive cellular immunotherapy, and photodynamic therapy after tail vein injection. These data show that the developed multifunctional nanostructure is a promising platform for efficient innate immunotherapy, photodynamic treatment and noninvasive therapeutic evaluation of breast cancer.


Asunto(s)
Neoplasias de la Mama , Antígeno CD56 , Células Asesinas Naturales , Fotoquimioterapia , Polietilenglicoles , Neoplasias de la Mama/terapia , Humanos , Femenino , Animales , Fotoquimioterapia/métodos , Ratones , Polietilenglicoles/química , Línea Celular Tumoral , Antígeno CD56/metabolismo , Inmunoterapia Adoptiva/métodos , Apoptosis/efectos de los fármacos , Imagen por Resonancia Magnética/métodos , Ratones Endogámicos BALB C , Ratones Desnudos
6.
J Nanobiotechnology ; 22(1): 7, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38166931

RESUMEN

Radionuclides internal radiotherapy (RIT) is a clinically powerful method for cancer treatment, but still poses unsatisfactory therapeutic outcomes due to the hypoxic characteristic of tumor microenvironment (TME). Catalase (CAT) or CAT-like nanomaterials can be used to enzymatically decompose TME endogenous H2O2 to boost TME oxygenation and thus alleviate the hypoxic level within tumors, but their effectiveness is still hindered by the short-lasting of hypoxia relief owing to their poor stability or degradability, thereby failing to match the long therapeutic duration of RIT. Herein, we proposed an innovative strategy of using facet-dependent CAT-like Pd-based two-dimensional (2D) nanoplatforms to continuously enhance RIT. Specifically, rationally designed 2D Pd@Au nanosheets (NSs) enable consistent enzymatic conversion of endogenous H2O2 into O2 to overcome hypoxia-induced RIT resistance. Furthermore, partially coated Au layer afford NIR-II responsiveness and moderate photothermal treatment that augmenting their enzymatic functionality. This approach with dual-effect paves the way for reshaping TME and consequently facilitating the brachytherapy ablation of cancer. Our work offers a significant advancement in the integration of catalytic nanomedicine and nuclear medicine, with the overarching goal of amplifying the clinical benefits of RIT-treated patients.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Peróxido de Hidrógeno , Microambiente Tumoral , Hipoxia/tratamiento farmacológico , Catálisis , Nanomedicina , Línea Celular Tumoral , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia
7.
J Nanobiotechnology ; 22(1): 101, 2024 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-38462598

RESUMEN

BACKGROUND: Radiotheranostics differs from the vast majority of other cancer therapies in its capacity for simultaneous imaging and therapy, and it is becoming more widely implemented. A balance between diagnostic and treatment requirements is essential for achieving effective radiotheranostics. Herein, we propose a proof-of-concept strategy aiming to address the profound differences in the specific requirements of the diagnosis and treatment of radiotheranostics. RESULTS: To validate the concept, we designed an s-tetrazine (Tz) conjugated prostate-specific membrane antigen (PSMA) ligand (DOTA-PSMA-Tz) for 68Ga or 177Lu radiolabeling and tumor radiotheranostics, a trans-cyclooctene (TCO) modified Pd@Au nanoplates (Pd@Au-PEG-TCO) for signal amplification, respectively. We then demonstrated this radiotheranostic strategy in the tumor-bearing mice with the following three-step procedures: (1) i.v. injection of the [68Ga]Ga-PSMA-Tz for diagnosis; (2) i.v. injection of the signal amplification module Pd@Au-PEG-TCO; (3) i.v. injection of the [177Lu]Lu-PSMA-Tz for therapy. Firstly, this strategy was demonstrated in 22Rv1 tumor-bearing mice via positron emission tomography (PET) imaging with [68Ga]Ga-PSMA-Tz. We observed significantly higher tumor uptake (11.5 ± 0.8%ID/g) with the injection of Pd@Au-PEG-TCO than with the injection [68Ga]Ga-PSMA-Tz alone (5.5 ± 0.9%ID/g). Furthermore, we validated this strategy through biodistribution studies of [177Lu]Lu-PSMA-Tz, with the injection of the signal amplification module, approximately five-fold higher tumor uptake of [177Lu]Lu-PSMA-Tz (24.33 ± 2.53% ID/g) was obtained when compared to [177Lu]Lu-PSMA-Tz alone (5.19 ± 0.26%ID/g) at 48 h post-injection. CONCLUSION: In summary, the proposed strategy has the potential to expand the toolbox of pretargeted radiotherapy in the field of theranostics.


Asunto(s)
Neoplasias Colorrectales , Radiofármacos , Masculino , Animales , Ratones , Radioisótopos de Galio , Distribución Tisular , Línea Celular Tumoral , Neoplasias Colorrectales/patología
8.
Med Sci Monit ; 30: e943526, 2024 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-38734884

RESUMEN

BACKGROUND A significant number of atrial fibrillation (AF) recurrences occur after initial ablation, often due to pulmonary vein reconnections or triggers from non-pulmonary veins. MATERIAL AND METHODS Patients with paroxysmal AF who underwent radiofrequency catheter ablation for the first time were enrolled. Base on propensity score matching (1: 1 matching), 118 patients were selected for an optimized workflow for the radiofrequency catheter ablation of paroxysmal AF (OWCA) group and a conventional group. Comparative analysis of the acute and 12-month clinical outcomes was conducted. Moreover, an artificial intelligence analytics platform was used to evaluate the quality of pulmonary vein isolation (PVI) circles. RESULTS PVI was successfully achieved in all patients. Incidence of first-pass isolation of bilateral PVI circles was higher (P=0.009) and acute pulmonary vein reconnections was lower (P=0.027) in the OWCA group than conventional group. The OWCA group displayed a significant reduction in the number of fractured points (P<0.001), stacked points (P=0.003), and a greater proportion of cases in which the radiofrequency index achieved the target value (P=0.003). Additionally, the contact force consistently met the force over time criteria (P<0.001) for bilateral PVI circles in the OWCA group, accompanied by a shorter operation time (P=0.017). During the 12-month follow-up period, the OWCA group exhibited a higher atrial arrhythmia-free survival rate following the initial ablation procedure than did the conventional group. CONCLUSIONS The optimized workflow for radiofrequency catheter ablation of paroxysmal AF could play a crucial role in creating higher quality PVI circles. This improvement is reflected in a significantly elevated 12-month atrial arrhythmia-free survival rate.


Asunto(s)
Fibrilación Atrial , Ablación por Catéter , Venas Pulmonares , Flujo de Trabajo , Humanos , Fibrilación Atrial/cirugía , Ablación por Catéter/métodos , Femenino , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Venas Pulmonares/cirugía , Anciano , Puntaje de Propensión , Recurrencia
9.
Nano Lett ; 23(16): 7699-7708, 2023 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-37565802

RESUMEN

Bone metastases are secondary malignant tumors that commonly occur after the spread of advanced cancer cells. We herein report the activatable semiconducting polymer nanoinducers (ASPNFP) that can amplify oxidative damage via sono-ferroptosis for bone metastasis treatment. ASPNFP are constructed by encapsulating plasma amine oxidase-based semiconducting polymer nanoparticles (SPNP) and Fe3O4 nanoparticles into singlet oxygen (1O2)-responsive nanocarriers. ASPNFP generate 1O2 under ultrasound (US) irradiation via a sonodynamic effect to destroy the stability of 1O2-responsive nanocarriers, allowing US-triggered releases of SPNP and Fe3O4 nanoparticles. SPNP decompose polyamines in tumor cells to produce acrolein and hydrogen peroxide (H2O2), in which H2O2 promotes Fenton reaction mediated by Fe3O4 nanoparticles for inducing enhanced ferroptosis and generation of hydroxyl radicals (•OH). The generated acrolein, 1O2, and •OH can simultaneously amplify the oxidative damage. ASPNFP thus mediate an amplified sono-ferroptosis effect to inhibit the growth of bone metastasis and restrict tumor metastasis.


Asunto(s)
Neoplasias Óseas , Ferroptosis , Nanopartículas , Neoplasias , Humanos , Acroleína , Peróxido de Hidrógeno , Neoplasias Óseas/tratamiento farmacológico , Estrés Oxidativo , Nanopartículas/uso terapéutico , Polímeros , Línea Celular Tumoral
10.
Angew Chem Int Ed Engl ; 63(2): e202310252, 2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38010197

RESUMEN

Bone metastasis is a type of metastatic tumors that involves the spreads of malignant tumor cells into skeleton, and its diagnosis and treatment remain a big challenge due to the unique tumor microenvironment. We herein develop osteoclast and tumor cell dual-targeting biomimetic semiconducting polymer nanocomposites (SPFeNOC ) for amplified theranostics of bone metastasis. SPFeNOC contain semiconducting polymer and iron oxide (Fe3 O4 ) nanoparticles inside core and surface camouflaged hybrid membrane of cancer cells and osteoclasts. The hybrid membrane camouflage enables their targeting to both metastatic tumor cells and osteoclasts in bone metastasis through homologous targeting mechanism, thus achieving an enhanced nanoparticle accumulation in tumors. The semiconducting polymer mediates near-infrared (NIR) fluorescence imaging and sonodynamic therapy (SDT), and Fe3 O4 nanoparticles are used for magnetic resonance (MR) imaging and chemodynamic therapy (CDT). Because both cancer cells and osteoclasts are killed synchronously via the combinational action of SDT and CDT, the vicious cycle in bone metastasis is broken to realize high antitumor efficacy. Therefore, 4T1 breast cancer-based bone metastasis can be effectively detected and cured by using SPFeNOC as dual-targeting theranostic nanoagents. This study provides an unusual biomimetic nanoplatform that simultaneously targets osteoclasts and cancer cells for amplified theranostics of bone metastasis.


Asunto(s)
Neoplasias Óseas , Nanocompuestos , Nanopartículas , Neoplasias , Humanos , Polímeros , Medicina de Precisión , Biomimética , Nanomedicina Teranóstica/métodos , Neoplasias Óseas/diagnóstico por imagen , Neoplasias Óseas/terapia , Nanocompuestos/uso terapéutico , Línea Celular Tumoral , Microambiente Tumoral
11.
Angew Chem Int Ed Engl ; 63(30): e202405639, 2024 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-38708791

RESUMEN

The high level of lactate in tumor microenvironment not only promotes tumor development and metastasis, but also induces immune escape, which often leads to failures of various tumor therapy strategies. We here report a sono-triggered cascade lactate depletion strategy by using semiconducting polymer nanoreactors (SPNLCu) for cancer cuproptosis-immunotherapy. The SPNLCu mainly contain a semiconducting polymer as sonosensitizer, lactate oxidase (LOx) conjugated via a reactive oxygen species (ROS)-cleavable linker and chelated Cu2+. Upon ultrasound (US) irradiation, the semiconducting polymer generates singlet oxygen (1O2) to cut ROS-cleavable linker to allow the release of LOx that catalyzes lactate depletion to produce hydrogen peroxide (H2O2). The Cu2+ will be reduced to Cu+ in tumor microenvironment, which reacts with the produced H2O2 to obtain hydroxyl radical (⋅OH) that further improves LOx release via destroying ROS-cleavable linkers. As such, sono-triggered cascade release of LOx achieves effective lactate depletion, thus relieving immunosuppressive roles of lactate. Moreover, the toxic Cu+ induces cuproptosis to cause immunogenic cell death (ICD) for activating antitumor immunological effect. SPNLCu are used to treat both subcutaneous and deep-tissue orthotopic pancreatic cancer with observably enhanced efficacy in restricting the tumor growths. This study thus provides a precise and effective lactate depletion tactic for cancer therapy.


Asunto(s)
Cobre , Inmunoterapia , Ácido Láctico , Neoplasias Pancreáticas , Polímeros , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/terapia , Ácido Láctico/química , Ácido Láctico/metabolismo , Polímeros/química , Polímeros/farmacología , Cobre/química , Humanos , Animales , Ratones , Semiconductores , Especies Reactivas de Oxígeno/metabolismo , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/química , Oxigenasas de Función Mixta/metabolismo , Oxigenasas de Función Mixta/química , Ondas Ultrasónicas , Línea Celular Tumoral , Antineoplásicos/química , Antineoplásicos/farmacología
12.
Angew Chem Int Ed Engl ; 63(30): e202401683, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-38719735

RESUMEN

Lanthanide nanoparticle (LnNP) scintillators exhibit huge potential in achieving radionuclide-activated luminescence (radioluminescence, RL). However, their structure-activity relationship remains largely unexplored. Herein, progressive optimization of LnNP scintillators is presented to unveil their structure-dependent RL property and enhance their RL output efficiency. Benefiting from the favorable host matrix and the luminescence-protective effect of core-shell engineering, NaGdF4 : 15 %Eu@NaLuF4 nanoparticle scintillators with tailored structures emerged as the top candidates. Living imaging experiments based on optimal LnNP scintillators validated the feasibility of laser-free continuous RL activated by clinical radiopharmaceuticals for tumor multiplex visualization. This research provides unprecedented insights into the rational design of LnNP scintillators, which would enable efficient energy conversion from Cerenkov luminescence, γ-radiation, and ß-electrons into visible photon signals, thus establishing a robust nanotechnology-aided approach for tumor-directed radio-phototheranostics.

13.
Biochem Biophys Res Commun ; 674: 27-35, 2023 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-37393641

RESUMEN

Intrinsic or acquired chemoresistance represents a major obstacle in cancer treatment. Multiple mechanisms can contribute to cancer cells' resistance to chemotherapy. Among them, an aberrantly strengthened DNA repair mechanism is responsible for a large proportion of drug resistance to alkylating agents and radiation therapy. In cancer cells, damping overactivated DNA repair system can overcome survival advantages conferred by chromosomal translocations or mutations and lead to cytostatic effects or cytotoxic. Therefore, selectively targeting DNA repair system in cancer cells holds promise for overcoming chemoresistance. In this study, we revealed that the endonuclease Flap Endonuclease 1 (FEN1), essential for DNA replication and repair, directly interacts with phosphatidylinositol 3-phosphate [PI(3)P], and FEN1-R378 is the primary PI(3)P-binding site. PI(3)P-binding deficient FEN1 mutant (FEN1-R378A) cells exhibited abnormal chromosomal structures and were hypersensitized to DNA damage. The PI(3)P-mediated FEN1 functionality was essential for repairing DNA damages caused by multiple mechanisms. Furthermore, VPS34, the major PI(3)P synthesizing enzyme, was negatively associated with patients' survival in various cancer types, and VPS34 inhibitors significantly sensitized chemoresistant cancer cells to genotoxic agents. These findings open up an avenue for counteracting chemoresistance by targeting VPS34-PI(3)P-mediated DNA repair pathway, and call for assessing the efficacy of this strategy in patients suffering from chemoresistance-mediated cancer recurrence in clinical trials.

14.
Bioconjug Chem ; 2023 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-36896731

RESUMEN

Regulation of gene expression is conducive to understanding the physiological roles of specific genes and provides therapeutic potentials, which however still remains a great challenge. Nonviral carriers have some advantages for gene delivery compared to traditional physical delivery strategies, but they often fail to control the delivery of genes in targeting regions, and thus lead to off-target side effects. Although endogenous biochemical signal-responsive carriers have been used to improve the transfection efficiency, their selectivity and specificity are still poor because of the coexistence of biochemical signals in both normal tissues and disease sites. In contrast, light-responsive carriers can be adopted to precisely control gene transgenic behaviors at the specified locations and time, thus reducing the off-target gene editing at nontarget positions. Particularly, the near-infrared (NIR) light has better tissue penetration depth and lower phototoxicity than ultraviolet and visible light sources, showing great promise for intracellular gene expression regulation. In this review, we summarize the recent progress of NIR photoresponsive nanotransducers for precision regulation of gene expression. These nanotransducers can achieve controlled gene expression via three different mechanisms (photothermal activation, photodynamic regulation, and NIR photoconversion) to allow various applications, such as gene therapy of cancer, which will be discussed in detail. A conclusion and discussion of the challenges and outlook will be given at the end of this review.

15.
Chemistry ; 29(17): e202203227, 2023 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-36484618

RESUMEN

The development of photothermal agents (PTAs) with robust photostability and high photothermal conversion efficiency is of great importance for cancer photothermal therapy. Herein, a novel PTA was created using two-dimensional intermetallic PtSnBi nanoplates (NPs), which demonstrated excellent photostability and biocompatibility with a high photothermal conversion efficiency of ∼61 % after PEGylation. More importantly, PtSnBi NPs could be employed as photoacoustic imaging contrast agents for tumor visualization due to their strong absorbance in the NIR range. In addition, both in vitro and in vivo experiments confirmed that PtSnBi NPs had a good photothermal efficacy under NIR laser irradiation. Therefore, the remarkable therapeutic characteristics of PtSnBi NPs make them a most promising candidate for cancer theranostics.


Asunto(s)
Nanopartículas , Neoplasias , Técnicas Fotoacústicas , Humanos , Fototerapia/métodos , Técnicas Fotoacústicas/métodos , Diagnóstico por Imagen , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Nanomedicina Teranóstica/métodos
16.
BMC Cardiovasc Disord ; 23(1): 466, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37715135

RESUMEN

BACKGROUND: Ethanol infusion of the vein of Marshall (EI-VOM) has been widely used to facilitate mitral isthmus (MI) ablation. According to the literature, the success rate of achieving a bidirectional conduction block across the MI ranges from 51 to 96%, with no standardized strategy or method available for cardiac electrophysiologists. OBJECTIVES: This study aimed to introduce and evaluate a novel ablation method of MI. METHODS: Consecutive patients with persistent atrial fibrillation (PeAF) that underwent catheter ablation were included. The MI ablation procedure followed a stepwise approach. In step 1, ethanol infusion of the vein of Marshall (EI-VOM) was performed. In step 2, a "V-shape" endocardial linear ablation connecting the left inferior pulmonary vein (LIPV) to mitral annulus (MA) was performed. In step 3, earliest activation sites(EASs) near the ablation line were identified using activation mapping followed by reinforced ablation. In step 4, precise epicardial ablation was performed, with the catheter introduced into the coronary sinus(CS) to target key ablation targets (KATs). RESULTS: 135 patients with PeAF underwent catheter ablation with the stepwise ablation method adopted in 119 cases. Bidirectional conduction blocks were achieved in 117 patients (98.3%). The block rates of every step were 0%, 58.0%, 44.0%, and 92.9%, and the cumulative block rates for the four steps were 0%, 58.0%, 76.5%, and 98.3%, respectively. No patient experienced fatal complications. CONCLUSIONS: Our novel stepwise catheter ablation method for MI yielded a high bidirectional block rate with high reproducibility.


Asunto(s)
Fibrilación Atrial , Ablación por Catéter , Humanos , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/cirugía , Reproducibilidad de los Resultados , Ablación por Catéter/efectos adversos , Catéteres , Etanol , Bloqueo Cardíaco , Válvula Mitral/diagnóstico por imagen , Válvula Mitral/cirugía
17.
Angew Chem Int Ed Engl ; 62(30): e202305200, 2023 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-37194682

RESUMEN

Immunotherapy has provided a promising modality for cancer treatment, while it often has the issues of limited response rates and potential off-target side effects in clinical practice. We herein report the construction of semiconducting polymer pro-nanomodulators (SPpMs) with ultrasound (US)-mediated activatable pharmacological actions for deep-tissue sono-immunotherapy of orthotopic pancreatic cancer. Such SPpMs consist of a sonodynamic semiconducting polymer backbone grafted with poly(ethylene glycol) chains linked with two immunomodulators (a programmed death-ligand 1 blocker and an indoleamine 2,3-dioxygenase inhibitor) via a singlet oxygen (1 O2 )-cleavable segment. In view of the excellent sonodynamic property of the semiconducting polymer core, SPpMs enable effective generation of 1 O2 under US treatment, even in a deep-tissue depth up to 12 cm. The generated 1 O2 not only ablates tumors via a sonodynamic effect and induces immunogenic cell death, but also destroys the 1 O2 -cleavable segments to allow in situ release of immunomodulators in tumors. This synergetic action results in boosted antitumor immune response via reversing two tumor immunosuppressive pathways. As such, SPpMs mediate deep-tissue sono-immunotherapy to completely eradicate orthotopic pancreatic cancer and effectively prevent tumor metastasis. Moreover, such an immune activation reduces the possibility of immune-related adverse events. This study thus provides a smart activatable nanoplatform for precise immunotherapy of deep-seated tumors.


Asunto(s)
Nanopartículas , Neoplasias , Neoplasias Pancreáticas , Humanos , Polímeros , Neoplasias/terapia , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Pancreáticas/tratamiento farmacológico , Polietilenglicoles , Inmunoterapia , Línea Celular Tumoral , Neoplasias Pancreáticas
18.
Eur J Nucl Med Mol Imaging ; 50(1): 27-37, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36066666

RESUMEN

PURPOSE: Stimulator of interferon genes (STING) protein plays a vital role in the immune surveillance of tumor microenvironment. Monitoring STING expression in tumors benefits the relevant STING therapy. This study aimed to develop a novel 18F-labeled agonist, dimeric amidobenzimidazole (diABZI), and firstly evaluate the feasibility of noninvasive positron emission tomography (PET) imaging of STING expression in the tumor microenvironment. METHODS: An analog of the STING agonist NOTA-DABI was synthesized and labeled with 18F via Al18F-NOTA complexation (denoted as [18F]F-DABI). Physicochemical properties, STING protein-binding affinity, and specificity of [18F]F-DABI were evaluated using cell uptake and docking assays. In vivo small-animal PET imaging and biodistribution studies of [18F]F-DABI in tumor-bearing mice were performed to verify the pharmacokinetics and tumor targeting ability. The correlation between tumor uptake and STING expression was also analyzed. RESULTS: [18F]F-DABI was produced conveniently with high radiochemical yield (44 ± 15%), radiochemical purity (> 97%) and molar activity (15-30 GBq/µmol). In vitro binding assays demonstrated that [18F]F-DABI has a favorable affinity and specificity for STING with a KD of 12.98 ± 2.07 nM. In vivo studies demonstrated the specificity of [18F]F-DABI for PET imaging of STING expression with B16F10 tumor uptake of 10.93 ± 0.93%ID/g, which was significantly different from that of blocking groups (3.13 ± 0.88%ID/g, ***p < 0.0001). Furthermore, tumor uptake of [18F]F-DABI was well positively correlated with STING expression in different tumor types. Biodistribution results demonstrated that [18F]F-DABI was predominately uptaken in the liver and intestines, indicating its hepatobiliary elimination. CONCLUSION: This proof-of-concept study demonstrated a STING-binding radioligand for PET imaging, which could be used as a potential companion diagnostic tool for related STING-agonist therapies.


Asunto(s)
Radioisótopos de Flúor , Tomografía de Emisión de Positrones , Animales , Ratones , Radioisótopos de Flúor/farmacocinética , Distribución Tisular , Línea Celular Tumoral , Tomografía de Emisión de Positrones/métodos , Expresión Génica , Interferones
19.
J Nanobiotechnology ; 20(1): 23, 2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-34991618

RESUMEN

Regulation of stimulator of interferon genes (STING) pathway using agonists can boost antitumor immunity for cancer treatment, while the rapid plasma clearance, limited membrane permeability, and inefficient cytosolic transport of STING agonists greatly compromise their therapeutic efficacy. In this study, we describe an extracellular matrix (ECM)-degrading nanoagonist (dNAc) with second near-infrared (NIR-II) light controlled activation of intracellular STING pathway for mild photothermal-augmented chemodynamic-immunotherapy of breast cancer. The dNAc consists of a thermal-responsive liposome inside loading with ferrous sulfide (FeS2) nanoparticles as both NIR-II photothermal converters and Fenton catalysts, 2'3'-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) as the STING agonist, and an ECM-degrading enzyme (bromelain) on the liposome surface. Mild heat generated by dNAc upon NIR-II photoirradiation improves Fenton reaction efficacy to kill tumor cells and cause immunogenic cell death (ICD). Meanwhile, the generated heat triggers a controlled release of cGAMP from thermal-responsive liposomes to active STING pathway. The mild photothermal activation of STING pathway combined with ICD promotes anti-tumor immune responses, which leads to improved infiltration of effector T cells into tumor tissues after bromelain-mediated ECM degradation. As a result, after treatment with dNAc upon NIR-II photoactivation, both primary and distant tumors in a murine mouse model are inhibited and the liver and lung metastasis are effectively suppressed. This work presents a photoactivatable system for STING pathway and combinational immunotherapy with improved therapeutic outcome.


Asunto(s)
Matriz Extracelular/metabolismo , Inmunoterapia , Proteínas de la Membrana , Nanopartículas , Fototerapia , Animales , Femenino , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Nanopartículas/metabolismo , Procesos Fotoquímicos
20.
Biochem Genet ; 60(5): 1511-1526, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35048221

RESUMEN

SnRK2 protein kinase family plays an important role in plant response to abiotic stress and has been identified in various plants. This study aimed to identify SnRK2 genes in tobacco and systematically analyze their expression under abscisic acid treatment and abiotic stress. We identified 22 NtSnRK2 members, which were divided into three groups and located on 13 chromosomes, mainly at both ends of the chromosomes; additionally, 11 duplicated NtSnRK2 gene pairs were observed. Phylogenetic analysis showed that these SnRK2 members were divided into three groups in tobacco. The motifs of NtSnRK2 proteins in the same group were highly similar. Subcellular localization indicated that NtSnRK2s in Group3 were present in the nucleus, cytomembrane, and cytoplasm. Gene expression pattern analysis revealed that NtSnRK2 genes played a role in the responses to several abiotic stresses (salt, drought, and low-temperature stress), indicating that they are widely involved in the adaptation of tobacco to adverse environmental conditions.


Asunto(s)
Regulación de la Expresión Génica de las Plantas , Nicotiana , Filogenia , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Estrés Fisiológico/genética , Nicotiana/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA