Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Nanobiotechnology ; 22(1): 234, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724978

RESUMEN

Radiotherapy-induced immune activation holds great promise for optimizing cancer treatment efficacy. Here, we describe a clinically used radiosensitizer hafnium oxide (HfO2) that was core coated with a MnO2 shell followed by a glucose oxidase (GOx) doping nanoplatform (HfO2@MnO2@GOx, HMG) to trigger ferroptosis adjuvant effects by glutathione depletion and reactive oxygen species production. This ferroptosis cascade potentiation further sensitized radiotherapy by enhancing DNA damage in 4T1 breast cancer tumor cells. The combination of HMG nanoparticles and radiotherapy effectively activated the damaged DNA and Mn2+-mediated cGAS-STING immune pathway in vitro and in vivo. This process had significant inhibitory effects on cancer progression and initiating an anticancer systemic immune response to prevent distant tumor recurrence and achieve long-lasting tumor suppression of both primary and distant tumors. Furthermore, the as-prepared HMG nanoparticles "turned on" spectral computed tomography (CT)/magnetic resonance dual-modality imaging signals, and demonstrated favorable contrast enhancement capabilities activated by under the GSH tumor microenvironment. This result highlighted the potential of nanoparticles as a theranostic nanoplatform for achieving molecular imaging guided tumor radiotherapy sensitization induced by synergistic immunotherapy.


Asunto(s)
Ferroptosis , Inmunoterapia , Compuestos de Manganeso , Proteínas de la Membrana , Ratones Endogámicos BALB C , Nanopartículas , Nucleotidiltransferasas , Óxidos , Fármacos Sensibilizantes a Radiaciones , Animales , Ratones , Inmunoterapia/métodos , Óxidos/química , Óxidos/farmacología , Femenino , Nucleotidiltransferasas/metabolismo , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Línea Celular Tumoral , Nanopartículas/química , Fármacos Sensibilizantes a Radiaciones/farmacología , Fármacos Sensibilizantes a Radiaciones/química , Proteínas de la Membrana/metabolismo , Ferroptosis/efectos de los fármacos , Glucosa Oxidasa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Humanos , Daño del ADN , Microambiente Tumoral/efectos de los fármacos
2.
J Exp Clin Cancer Res ; 43(1): 62, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38419081

RESUMEN

BACKGROUND: In recent years, the development of adjunctive therapeutic hyperthermia for cancer therapy has received considerable attention. However, the mechanisms underlying hyperthermia resistance are still poorly understood. In this study, we investigated the roles of cold­inducible RNA binding protein (Cirbp) in regulating hyperthermia resistance and underlying mechanisms in nasopharyngeal carcinoma (NPC). METHODS: CCK-8 assay, colony formation assay, tumor sphere formation assay, qRT-PCR, Western blot were employed to examine the effects of hyperthermia (HT), HT + oridonin(Ori) or HT + radiotherapy (RT) on the proliferation and stemness of NPC cells. RNA sequencing was applied to gain differentially expressed genes upon hyperthermia. Gain-of-function and loss-of-function experiments were used to evaluate the effects of RNAi-mediated Cirbp silencing or Cirbp overexpression on the sensitivity or resistance of NPC cells and cancer stem-like cells to hyperthermia by CCK-8 assay, colony formation assay, tumorsphere formation assay and apoptosis assay, and in subcutaneous xenograft animal model. miRNA transient transfection and luciferase reporter assay were used to demonstrate that Cirbp is a direct target of miR-377-3p. The phosphorylation levels of key members in ATM-Chk2 and ATR-Chk1 pathways were detected by Western blot. RESULTS: Our results firstly revealed that hyperthermia significantly attenuated the stemness of NPC cells, while combination treatment of hyperthermia and oridonin dramatically increased the killing effect on NPC cells and cancer stem cell (CSC)­like population. Moreover, hyperthermia substantially improved the sensitivity of radiation­resistant NPC cells and CSC­like cells to radiotherapy. Hyperthermia noticeably suppressed Cirbp expression in NPC cells and xenograft tumor tissues. Furthermore, Cirbp inhibition remarkably boosted anti­tumor­killing activity of hyperthermia against NPC cells and CSC­like cells, whereas ectopic expression of Cirbp compromised tumor­killing effect of hyperthermia on these cells, indicating that Cirbp overexpression induces hyperthermia resistance. ThermomiR-377-3p improved the sensitivity of NPC cells and CSC­like cells to hyperthermia in vitro by directly suppressing Cirbp expression. More importantly, our results displayed the significantly boosted sensitization of tumor xenografts to hyperthermia by Cirbp silencing in vivo, but ectopic expression of Cirbp almost completely counteracted hyperthermia-mediated tumor cell-killing effect against tumor xenografts in vivo. Mechanistically, Cirbp silencing-induced inhibition of DNA damage repair by inactivating ATM-Chk2 and ATR-Chk1 pathways, decrease in stemness and increase in cell death contributed to hyperthermic sensitization; conversely, Cirbp overexpression-induced promotion of DNA damage repair, increase in stemness and decrease in cell apoptosis contributed to hyperthermia resistance. CONCLUSION: Taken together, these findings reveal a previously unrecognized role for Cirbp in positively regulating hyperthermia resistance and suggest that thermomiR-377-3p and its target gene Cirbp represent promising targets for therapeutic hyperthermia.


Asunto(s)
Diterpenos de Tipo Kaurano , Hipertermia Inducida , MicroARNs , Neoplasias Nasofaríngeas , Animales , Humanos , Neoplasias Nasofaríngeas/patología , Sincalida/metabolismo , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/terapia , Carcinoma Nasofaríngeo/patología , MicroARNs/genética , Células Madre Neoplásicas/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica
3.
Adv Healthc Mater ; : e2303626, 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38387885

RESUMEN

Immunotherapy has emerged as an innovative strategy with the potential to improve outcomes in cancer patients. Recent evidence indicates that radiation-induced DNA damage can activate the cyclic-GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway to enhance the antitumor immune response. Even so, only a small fraction of patients currently benefits from radioimmunotherapy due to the radioresistance and the inadequate activation of the cGAS-STING pathway. Herein, this work integrates hafnium oxide (HfO2 ) nanoparticles (radiosensitizer) and 7-Ethyl-10-hydroxycamptothecin (SN38, chemotherapy drug, STING agonist) into a polydopamine (PDA)-coated core-shell nanoplatform (HfO2 @PDA/Fe/SN38) to achieve synergistic chemoradiotherapy and immunotherapy. The co-delivery of HfO2 /SN38 greatly enhances radiotherapy efficacy by effectively activating the cGAS-STING pathway, which then triggers dendritic cells maturation and CD8+ T cells recruitment. Consequently, the growth of both primary and abscopal tumors in tumor-bearing mice is efficiently inhibited. Moreover, the HfO2 @PDA/Fe/SN38 complexes exhibit favorable magnetic resonance imaging (MRI)/photoacoustic (PA) bimodal molecular imaging properties. In summary, these developed multifunctional complexes have the potential to intensify immune activation to realize simultaneous cancer Radio/Chemo/Immunotherapy for clinical translation.

4.
Small ; 19(43): e2302758, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37381095

RESUMEN

Innate immunity triggered by the cGAS/STING pathway has the potential to improve cancer immunotherapy. Previously, the authors reported that double-stranded DNA (dsDNA) released by dying tumor cells can trigger the cGAS/STING pathway. However, owing to efferocytosis, dying tumor cells are engulfed and cleared before the damaged dsDNA is released; hence, immunologic tolerance and immune escape occur. Herein, a cancer-cell-membrane biomimetic nanocomposites that exhibit tumor-immunotherapeutic effects are synthesized by augmenting the cGAS/STING pathway and suppressing efferocytosis. Once internalized by cancer cells, a combined chemo/chemodynamic therapy would be triggered, which damages their nuclear and mitochondrial DNA. Furthermore, the releasing Annexin A5 protein could inhibit efferocytosis effect and promote immunostimulatory secondary necrosis by preventing phosphatidylserine exposure, resulting in the burst release of dsDNA. These dsDNA fragments, as molecular patterns to immunogenic damage, escape from the cancer cells, activate the cGAS/STING pathway, enhance cross-presentation inside dendritic cells, and promote M1-polarization of tumor-associated macrophages. In vivo experiments suggest that the proposed nanocomposite could recruit cytotoxic T-cells and facilitate long-term immunological memory. Moreover, when combined with immune-checkpoint blockades, it could augment the immune response. Therefore, this novel biomimetic nanocomposite is a promising strategy for generating adaptive antitumor immune responses.


Asunto(s)
Proteínas de la Membrana , Neoplasias , Humanos , Proteínas de la Membrana/metabolismo , Inmunidad Innata , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Neoplasias/terapia , ADN , Membrana Celular/metabolismo , Inmunoterapia/métodos
5.
Acta Biomater ; 162: 72-84, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36931419

RESUMEN

Although radiotherapeutic efficiency has been revealed to be positively correlated with ferroptosis, the neutral/alkaline cytoplasm pH value of tumor cells remains an intrinsic challenge for efficient Fenton/Fenton-like reaction-based ferroptosis induction. Herein, PEGylated hollow mesoporous organosilica nanotheranostics (HMON)-GOx@MnO2 nanoparticles (HGMP NPs) were designed as a ferroptosis inducer, which could specifically release Mn2+ in tumor cells to activate the Fenton-like reaction for ferroptosis induction. Proton pump inhibitors (PPIs) were synchronously administered for cytoplasm pH level regulation by inhibiting V-H+-ATPases activity, enhancing Fenton-like reaction-based ferroptosis induction. Moreover, reactive oxygen species production was facilitated via the glucose oxidase triggered cascade catalytic reaction by utilizing intracellular ß-D-glucose for H2O2 self-supply and generation of additional cytoplasm H+. The PPI enhanced ferroptosis inducing nanosystem effectively inhibited tumor growth both in vitro and in vivo for tumor-specific ferroptosis induction and radiotherapy sensitization, suggesting that PPI administration could be an efficient adjuvant to reinforce Fenton/Fenton-like reaction-based ferroptosis induction for radiosensitization. STATEMENT OF SIGNIFICANCE: The cytoplasm pH value of tumor cells is typically neutral to alkaline, which is higher than that of the Fenton/Fenton-like reaction desired acidic environments, hindering its efficiency. In this study, PEGylated hollow mesoporous organosilica nanotheranostics (HMON)-GOx@MnO2 nanoparticles were synthesized as a ferroptosis inducer, which could specifically release Mn2+ via depleting glutathione and then activate the Fenton-like reaction in the tumor microenvironment. The glucose oxidase was applied for H2O2 self-supply and addition of cytoplasm H+ to further boost the Fenton-like reaction. We found that proton pump inhibitors (PPIs) increased intracellular acidification by inhibiting the activity of V-H+-ATPases to enhance the Fenton reaction-based ferroptosis induction, suggesting PPIs administration could be a feasible strategy to reinforce ferroptosis induction for radiosensitization.


Asunto(s)
Ferroptosis , Nanopartículas , Neoplasias , Humanos , Inhibidores de la Bomba de Protones , Glucosa Oxidasa , Peróxido de Hidrógeno/farmacología , Compuestos de Manganeso/farmacología , Óxidos , Polietilenglicoles , Adenosina Trifosfatasas , Línea Celular Tumoral , Microambiente Tumoral
6.
Oxid Med Cell Longev ; 2022: 4674215, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36111165

RESUMEN

Lipotoxicity can lead to beta-cell dysfunction and apoptosis because it induces oxidative stress. Recent studies have found that Irisin prevents pancreatic beta-cell dysfunction induced by palmitic acid (PA). However, an association between the protection against oxidative stress conferred by Irisin and beta-cell dysfunction has not been fully elucidated. In this study, we observed that Irisin treatment prevented INS-1 cell apoptosis induced by PA treatment and preserved the insulin-secreting function of INS-1 cells in vitro. These effects probably resulted from the Irisin-induced decrease in intracellular ROS levels triggered by PA treatment. In addition, PA treatment induced oxidative stress partially by inhibiting the activation of thioredoxin 2 (Trx2) through its increase of thioredoxin-interacting protein (Txnip) expression. However, Irisin administration blocked the increase in Txnip expression, which reversed the PA-induced inactivation of Trx2. Irisin also increased the nuclear translocation of Stat3, and the inhibition of Stat3 by siRNAs blocked Irisin-induced Trx2 expression, indicating that both Txnip and Stat3 are involved in Irisin-induced activation of Trx2. Furthermore, blockade of Stat3 by siRNAs led to the decreased gene expression of MafA and Ins and to cessation of glucose-induced insulin secretion that had been enhanced by Irisin. In vivo, HFD treatment led to reduced glucose tolerance and an increase in the level of the oxidative marker malondialdehyde (MDA) compared to that in the control group. However, these effects were ameliorated by Irisin injection due to the inhibition of beta-cell apoptosis and the activation of Trx2, probably through Txnip inhibition and Stat3 activation. In conclusion, our results reveal a possible mechanism for Irisin-induced beta-cell protection, which is mediated through Txnip inhibition and activation of the Stat3-Trx2 pathway.


Asunto(s)
Fibronectinas , Tiorredoxinas , Fibronectinas/metabolismo , Glucosa/toxicidad , Insulina/metabolismo , Malondialdehído/metabolismo , Estrés Oxidativo , Ácido Palmítico/toxicidad , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/farmacología , Tiorredoxinas/metabolismo
7.
J Control Release ; 347: 369-378, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35577149

RESUMEN

Chemo-radiotherapy has been extensively used in clinics, displaying substantial advantages in treatment and prognosis. Stimuli-responsive biodegradable nanoagents that can achieve not only delivery and controlled release of chemotherapeutics, but also hypoxia alleviation to enhance chemoradiotherapy therefore has tremendous potential. Herein, glutathione (GSH)-responsive, biodegradable, doxorubicin-carrying hollow mesoporous organotantalum nanospheres modified with Au and Pt dual nanoenzymes (HMOTP@Pt@Au@Dox) were constructed for chemo-radio sensitization. Degradation of HMOTP@Pt@Au@Dox can be self-activated through GSH stimulation and on-demand release packaged Dox owing to the disulfide bond in the hybrid framework of organotantalum nanospheres. Au and Pt nanoenzymes triggered cascade catalytic reactions that could alleviate hypoxia by utilizing ß-d-glucose and H2O2, thereby sensitizing ROS-based chemoradiotherapy with synergistic starving therapy. Given the radiosensitization of high-Z elements (Ta, Pt, Au), nanoenzymes induced cascade catalytic reaction for hypoxia relief, and the depletion of the predominant antioxidant GSH, desirable tumor suppression could be achieved both in vitro and in vivo, indicating that HMOTP@Pt@Au@Dox is a promising nanoagent to boost chemo-radiotherapy.


Asunto(s)
Nanopartículas , Nanosferas , Línea Celular Tumoral , Doxorrubicina , Glutatión/metabolismo , Humanos , Peróxido de Hidrógeno , Hipoxia , Nanopartículas/química
8.
J Control Release ; 347: 55-67, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35489546

RESUMEN

Though amounts of attempts about nanomedicine for chemo-radiotherapy have been made, more efficient strategies for chemo-radio therapy enhancement still need to be studied and perfected. Herein, a 'yolk-shell'-like nanostructure (Bi2S3@mBixMnyOz nanosystem) was facilely constructed by directly using radiosensitizer Bi2S3 nanorods (NRs) as a partial sacrificial template. Then, the chemotherapeutic drug doxorubicin (DOX) loaded PEGylated Bi2S3@mBixMnyOz nanosystem (PBmB-DOX) was constructed, which could realize tumor microenvironment (TME)-responsive drug release for chemotherapy sensitivity enhancement. And the Bi2S3 NRs core could deposit more radiant energy to improve the radiotherapy sensitivity. Meanwhile, the compounds shell could catalyze H2O2 to generate O2, so as to alleviate tumor hypoxia for further chemo-radio therapy sensitization enhancement. More importantly, ferroptosis was participated in the process of PBmB-induced therapy via glutathione (GSH)-depletion mediated GPX4 inactivation, together with Mn ions induced chemodynamic therapy (Fenton-like reaction), which made additional contributions to increase the therapeutic efficacy. Last but not least, the GSH-stimulated degradation of compounds shell could contribute to self-enhanced T1-MR imaging activation, which allowed on-demand tumor diagnosis. In this work, the synthetic strategy that directly using Bi2S3 NRs as a partial sacrificial template to rapidly synthesize the 'yolk-shell'-like nanostructure for nanomedical application has rarely been reported before. And the in vitro and in vivo results suggest that our 'yolk-shell'-like PBmB-DOX nanosystem holds great promise to regulate TME for tumor-specific diagnosis and synergistic therapy.


Asunto(s)
Nanopartículas , Neoplasias , Línea Celular Tumoral , Doxorrubicina/química , Glutatión/metabolismo , Humanos , Peróxido de Hidrógeno , Imagen por Resonancia Magnética , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Hipoxia Tumoral , Microambiente Tumoral
9.
Int J Nanomedicine ; 17: 395-407, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35115774

RESUMEN

BACKGROUND: Radiotherapy (RT) is clinically well-established cancer treatment. However, radioresistance remains a significant issue associated with failure of RT. Phototherapy-induced radiosensitization has recently attracted attention in translational cancer research. METHODS: Cu-Sb-S nanoparticles (NPs) coated with ultra-small Au nanocrystals (Au@Cu-Sb-S) were synthesized and characterized. The biosafety profiles, absorption of near-infrared (NIR) laser and radiation-enhancing effect of the NPs were evaluated. In vitro and in vivo spectral computed tomography (CT) imaging and photoacoustic (PA) imaging were performed in 4T1 breast cancer-bearing mice. The synergetic radio-phototherapy was assessed by in vivo tumor inhibition studies. RESULTS: Au@Cu-Sb-S NPs were prepared by in situ growth of Au NCs on the surface of Cu-Sb-S NPs. The cell viability experiments showed that the combination of Au@Cu-Sb-S+NIR+RT was significantly more cytotoxic to tumor cells than the other treatments at concentrations above 25 ppm Sb. In vitro and in vivo spectral CT imaging demonstrated that the X-ray attenuation ability of Au@Cu-Sb-S NPs was superior to that of the clinically used Iodine, particularly at lower KeV levels. Au@Cu-Sb-S NPs showed a concentration-dependent and remarkable PA signal brightening effect. In vivo tumor inhibition studies showed that the prepared Au@Cu-Sb-S NPs significantly suppressed tumor growth in 4T1 breast cancer-bearing mice treated with NIR laser irradiation and an intermediate X-ray dose (4 Gy). CONCLUSION: These results indicate that Au@Cu-Sb-S integrated with spectral CT, PA imaging, and phototherapy-enhanced radiosensitization is a promising multifunctional theranostic nanoplatform for clinical applications.


Asunto(s)
Hipertermia Inducida , Nanopartículas , Neoplasias , Técnicas Fotoacústicas , Animales , Línea Celular Tumoral , Ratones , Fototerapia , Nanomedicina Teranóstica , Tomografía Computarizada por Rayos X
10.
Acta Pharm Sin B ; 11(10): 3231-3243, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34729312

RESUMEN

Ferroptosis, as a newly discovered cell death form, has become an attractive target for precision cancer therapy. Several ferroptosis therapy strategies based on nanotechnology have been reported by either increasing intracellular iron levels or by inhibition of glutathione (GSH)-dependent lipid hydroperoxidase glutathione peroxidase 4 (GPX4). However, the strategy by simultaneous iron delivery and GPX4 inhibition has rarely been reported. Herein, novel tumor microenvironments (TME)-activated metal-organic frameworks involving Fe & Cu ions bridged by disulfide bonds with PEGylation (FCSP MOFs) were developed, which would be degraded specifically under the redox TME, simultaneously achieving GSH-depletion induced GPX4 inactivation and releasing Fe ions to produce ROS via Fenton reaction, therefore causing ferroptosis. More ROS could be generated by the acceleration of Fenton reaction due to the released Cu ions and the intrinsic photothermal capability of FCSP MOFs. The overexpressed GSH and H2O2 in TME could ensure the specific TME self-activated therapy. Better tumor therapeutic efficiency could be achieved by doxorubicin (DOX) loading since it can not only cause apoptosis, but also indirectly produce H2O2 to amplify Fenton reaction. Remarkable anti-tumor effect of obtained FCSP@DOX MOFs was verified via both in vitro and in vivo assays.

11.
J Nanobiotechnology ; 19(1): 146, 2021 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-34011375

RESUMEN

BACKGROUND: Paclitaxel (PTX) has been suggested to be a promising front-line drug for gastric cancer (GC), while P-glycoprotein (P-gp) could lead to drug resistance by pumping PTX out of GC cells. Consequently, it might be a hopeful way to combat drug resistance by inhibiting the out-pumping function of P-gp. RESULTS: In this study, we developed a drug delivery system incorporating PTX onto polyethylene glycol (PEG)-modified and oxidized sodium alginate (OSA)-functionalized graphene oxide (GO) nanosheets (NSs), called PTX@GO-PEG-OSA. Owing to pH/thermal-sensitive drug release properties, PTX@GO-PEG-OSA could induced more obvious antitumor effects on GC, compared to free PTX. With near infrared (NIR)-irradiation, PTX@GO-PEG-OSA could generate excessive reactive oxygen species (ROS), attack mitochondrial respiratory chain complex enzyme, reduce adenosine-triphosphate (ATP) supplement for P-gp, and effectively inhibit P-gp's efflux pump function. Since that, PTX@GO-PEG-OSA achieved better therapeutic effect on PTX-resistant GC without evident toxicity. CONCLUSIONS: In conclusion, PTX@GO-PEG-OSA could serve as a desirable strategy to reverse PTX's resistance, combined with chemo/photothermal/photodynamic therapy.


Asunto(s)
Adenosina Trifosfato/metabolismo , Grafito/química , Grafito/farmacología , Mitocondrias/efectos de los fármacos , Paclitaxel/farmacología , Fotoquimioterapia/métodos , Neoplasias Gástricas/tratamiento farmacológico , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones , Ratones Desnudos , Mitocondrias/metabolismo , Nanopartículas/química , Nanopartículas/uso terapéutico , Fototerapia , Polietilenglicoles , Células RAW 264.7 , Especies Reactivas de Oxígeno
12.
Front Bioeng Biotechnol ; 9: 635858, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33681171

RESUMEN

Melanin, as a natural product, has been used as an extraordinary ingredient for nanomedicine due to its great biocompatibility and light responsive property. In this study, polydopamine (PDA), an analog of melanin, was extracted from dopamine and encapsulated with doxorubicin (DOX). The as-prepared nanoparticles (NPs) with good stability, great biosafety and high near infrared (NIR) responsive property ameliorated the cell uptake of DOX in OS-RC-2/ADR cells, exhibited synergistic chemo/photothermal (PTT)/photodynamic (PDT) effects, induced the release of damage associated molecular patterns (DAMPs), and finally, led to immunogenic cell death (ICD). In general, it was suggested that PDA-DOX NPs with NIR irradiation could serve as a promising agent for tumor therapy.

13.
Mater Sci Eng C Mater Biol Appl ; 120: 111722, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33545873

RESUMEN

Among various methods, the use of targeting nucleic acid therapy is a promising method for inhibiting gastric cancer (GC) cells' rapid growth and metastasis abilities. In this study, vitamin B12-labeled poly (d,l-lactide-co-glycolide) and polyethylene glycol nanoparticles (PLGA-PEG-VB12 NPs) were developed for microRNAs-532-3p mimics incorporating as targeting gene delivery systems (miR-532-3p@PLGA-PEG-VB12 NPs) to fight against transcobalamin II (CD320)-overexpressed GC cells' progression. The PLGA-PEG-VB12 NPs with appropriate particle sizes and good bio-compatibility could be selectively delivered into CD320-overexpressed GC cells, and significantly decrease the expression of apoptosis repressor with caspase recruitment domain (ARC). Following that, more pro-apoptotic protein (Bax) flowed from cytoplasm into mitochondria to form Bax oligomerization, thus induced mitochondrial damage, including mitochondrial membrane potentials (MMPs) loss and excessive production of mitochondrial reactive oxygen species (mitoROS). Since that, mitochondrial permeability transition pore (mPTP) was opened, followed by induced more cytochrome c (Cyto C) releasing from mitochondria into cytosol, and finally activated caspase-depended cell apoptosis pathway. Therefore, our designed miR-532-3p@PLGA-PEG-VB12 NPs showed enhanced GC targeting ability, and could induce apoptosis through activating ARC/Bax/mitochondria-mediated apoptosis signaling pathway, finally remarkably suppressed proliferation of GC cells both in vitro and in vivo, which presented a promising treatment for GC.


Asunto(s)
MicroARNs , Nanopartículas , Neoplasias Gástricas , Antígenos CD , Apoptosis , Dominio de Reclutamiento y Activación de Caspasas , Humanos , MicroARNs/genética , Polietilenglicoles , Receptores de Superficie Celular , Neoplasias Gástricas/tratamiento farmacológico , Vitamina B 12 , Vitaminas
14.
J Nanobiotechnology ; 18(1): 99, 2020 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-32690085

RESUMEN

BACKGROUND: CuS-modified hollow mesoporous organosilica nanoparticles (HMON@CuS) have been preferred as non-invasive treatment for cancer, as near infrared (NIR)-induced photo-thermal effect (PTT) and/or photo-dynamic effect (PDT) could increase cancer cells' apoptosis. However, the certain role of HMON@CuS-produced-PTT&PDT inducing gastric cancer (GC) cells' mitochondrial damage, remained unclear. Moreover, theranostic efficiency of HMON@CuS might be well improved by applying multi-modal imaging, which could offer an optimal therapeutic region and time window. Herein, new nanotheranostics agents were reported by Gd doped HMON decorated by CuS nanocrystals (called HMON@CuS/Gd). RESULTS: HMON@CuS/Gd exhibited appropriate size distribution, good biocompatibility, L-Glutathione (GSH) responsive degradable properties, high photo-thermal conversion efficiency (82.4%) and a simultaneous reactive oxygen species (ROS) generation effect. Meanwhile, HMON@CuS/Gd could efficiently enter GC cells, induce combined mild PTT (43-45 °C) and PDT under mild NIR power density (0.8 W/cm2). Surprisingly, it was found that PTT might not be the only factor of cell apoptosis, as ROS induced by PDT also seemed playing an essential role. The NIR-induced ROS could attack mitochondrial transmembrane potentials (MTPs), then promote mitochondrial reactive oxygen species (mitoROS) production. Meanwhile, mitochondrial damage dramatically changed the expression of anti-apoptotic protein (Bcl-2) and pro-apoptotic protein (Bax). Since that, mitochondrial permeability transition pore (mPTP) was opened, followed by inducing more cytochrome c (Cyto C) releasing from mitochondria into cytosol, and finally activated caspase-9/caspase-3-depended cell apoptosis pathway. Our in vivo data also showed that HMON@CuS/Gd exhibited good fluorescence (FL) imaging (wrapping fluorescent agent), enhanced T1 imaging under magnetic resonance imaging (MRI) and infrared thermal (IRT) imaging capacities. Guided by FL/MRI/IRT trimodal imaging, HMON@CuS/Gd could selectively cause mild photo-therapy at cancer region, efficiently inhibit the growth of GC cells without evident systemic toxicity in vivo. CONCLUSION: HMON@CuS/Gd could serve as a promising multifunctional nanotheranostic platform and as a cancer photo-therapy agent through inducing mitochondrial dysfunction on GC.


Asunto(s)
Mitocondrias , Imagen Multimodal/métodos , Compuestos de Organosilicio , Fototerapia/métodos , Neoplasias Gástricas , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sulfato de Cobre , Humanos , Imagen por Resonancia Magnética , Mitocondrias/patología , Mitocondrias/efectos de la radiación , Compuestos de Organosilicio/química , Compuestos de Organosilicio/farmacología , Neoplasias Gástricas/diagnóstico por imagen , Neoplasias Gástricas/patología , Neoplasias Gástricas/terapia , Nanomedicina Teranóstica
15.
Nanotoxicology ; 14(6): 774-787, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32401088

RESUMEN

Gastric carcinoma is one of the most lethal malignant tumors. As part of our long-term efforts on seeking effective diagnosis and therapeutic strategies of gastric cancer, we present herein novel ternary copper-based chalcogenide nanoplatform CuS-NiS2 nanomaterials with outstanding photothermal (PT)/photodynamic (PD) property that could effectively suppress human gastric cancer in vitro and in vivo without obvious side effects. We revealed that CuS-NiS2 induced reactive oxygen species (ROS) generation, leading to apoptosis through Bcl-2/Bax pathway of human gastric cancer cells under 808 nm near-infrared (NIR) irradiation. In addition, we also confirmed that the combination of CuS-NiS2 and 808 nm NIR laser treatment triggered necroptosis by regulating the novel pathway MLKL/CAPG of human gastric cancer cells. Moreover, the CuS-NiS2 exhibited excellent contrast enhancement according to magnetic resonance imaging (MRI). Taken together, we reported new ternary copper-based chalcogenide nanomaterials CuS-NiS2, which could be successfully applied for MRI-guided PT/PD therapy of gastric carcinoma through mitochondria-mediated apoptosis and MLKL/CAPG-mediated necroptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Cobre/uso terapéutico , Proteínas de Microfilamentos/metabolismo , Mitocondrias/efectos de los fármacos , Nanoestructuras/uso terapéutico , Necroptosis/efectos de los fármacos , Níquel/uso terapéutico , Proteínas Nucleares/metabolismo , Fototerapia/métodos , Proteínas Quinasas/metabolismo , Neoplasias Gástricas/terapia , Animales , Línea Celular Tumoral , Cobre/administración & dosificación , Humanos , Imagen por Resonancia Magnética , Masculino , Ratones Desnudos , Mitocondrias/metabolismo , Nanoestructuras/administración & dosificación , Níquel/administración & dosificación , Neoplasias Gástricas/diagnóstico por imagen , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Biomater Sci ; 8(8): 2202-2211, 2020 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-32100739

RESUMEN

The efficiency of drug delivery and bioavailability to tumor cells are crucial for effective cancer chemotherapy. Herein, a doxorubicin (DOX) encapsulated lysolipid-based thermosensitive liposome decorated with cRGD peptide (RTSL) is conjugated on the surface of an IR780-loaded microbubble (IMB) to synthesize RTSL-IMBs. Sequentially taking advantage of acoustic-assisted early extravasation and thermo-triggered interstitium ultrafast drug release, RTSL-IMBs combine with ultrasound (US) and laser irradiation can advance drug delivery and bioavailability. In vitro experiments demonstrate that RTSL-IMBs associated with a two-step protocol (subsequently US irradiation for 1 min and laser irradiation for 5 min) can dramatically enhance the cellular uptake and bioavailability of DOX. In vivo fluorescence imaging studies reveal that the combination of RTSL-IMBs and US shows a 2.8-fold intratumoral drug accumulation increase at 0.5 h post-injection, while it will take 48 h to reach the same level of intratumoral drug accumulation for the RTSL-IMB group alone. Interestingly, the following localized application of a laser can further increase drug accumulation and slow tumor clearance. Histological analysis demonstrates that the combinational RTSL-IMBs, US and laser significantly improve the drug penetration distance and delivery efficiency in the tumor core. In this study, the acoustic/thermo-responsive hybrid system shows potential for advancing DOX chemotherapy in breast cancer cell MCF-7 xenograft nude mice.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Péptidos Cíclicos/administración & dosificación , Acústica , Animales , Antibióticos Antineoplásicos/química , Doxorrubicina/química , Liberación de Fármacos , Femenino , Humanos , Liposomas , Células MCF-7 , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones Endogámicos BALB C , Ratones Desnudos , Microburbujas , Péptidos Cíclicos/química , Temperatura , Carga Tumoral/efectos de los fármacos , Ondas Ultrasónicas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
ACS Appl Mater Interfaces ; 12(5): 5520-5530, 2020 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-31891473

RESUMEN

Radiotherapy (RT) is one of the most widely used cancer treatments in the clinical setting, while hypoxia-associated resistance often occurs. Herein, a PEGylated TaOx-based oxygen-carrying nanoplatform was constructed for triple sensitizing tumor radiotherapy. The high-Z element based hollow mesoporous TaOx nanospheres were prepared following the in situ growth of ultrasmall CuS nanocrystals and then packaged with O2-saturated perfluoropentane (PFP). NIR laser-triggered mild hyperthermia would lead to the increase of intratumoral blood flow, together with the release of O2, the radiotherapeutic efficiency would be enhanced. Alternatively, radiant energy would be deposited inside the tumor by the Ta element, therefore triple sensitization of radiotherapy could be achieved. The in vivo studies showed that the as-prepared nanospheres could achieve almost total inhibition of tumor growth without obvious side effects, which provides new possibilities for multisensitizing tumor radiotherapy.


Asunto(s)
Materiales Biocompatibles/uso terapéutico , Nanosferas/química , Neoplasias/terapia , Óxidos/química , Tantalio/química , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cobre/química , Femenino , Fluorocarburos/química , Humanos , Hipertermia Inducida , Rayos Infrarrojos/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Nanosferas/toxicidad , Neoplasias/patología , Neoplasias/radioterapia , Oxígeno/química , Porosidad , Especies Reactivas de Oxígeno/metabolismo
18.
Nanoscale ; 11(27): 12983-12989, 2019 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-31264665

RESUMEN

With a high incidence and high mortality rate, ovarian cancer presents a challenge for clinical practice. It is thus extremely urgent to investigate new diagnosis and therapy methods for the treatment of ovarian cancer. Ternary copper-based chalcogenide nanomaterials are attractive owing to their near infrared (NIR) response for cancer theranostic fields. However, improving the theranostic efficiency of these nanomaterials is challenging. Herein, CuS-MnS2 nano-flowers were easily synthesized and under NIR irradiation exhibited a relatively high photothermal conversion efficiency of 67.5% and a simultaneous reactive oxygen species (ROS) generation effect. Owing to these outstanding photothermal/photodynamic effects, excellent tumor ablation results could be achieved by the combined use of CuS-MnS2 nano-flowers and 808 nm NIR laser treatments. The main anticancer mechanism of CuS-MnS2 nano-flowers + NIR was likely necroptosis. In addition, the nano-flowers showed remarkable contrast enhancement according to magnetic resonance imaging (MRI). These CuS-MnS2 nano-flowers could thus serve as a promising multifunctional nanotheranostic agent for MRI and as a photothermal/photodynamic cancer therapy agent through necroptosis.


Asunto(s)
Cobre , Hipertermia Inducida , Imagen por Resonancia Magnética , Compuestos de Manganeso , Nanopartículas , Necroptosis/efectos de los fármacos , Neoplasias Experimentales , Neoplasias Ováricas , Fototerapia , Sulfuros , Células A549 , Animales , Cobre/química , Cobre/farmacología , Femenino , Humanos , Compuestos de Manganeso/química , Compuestos de Manganeso/farmacología , Ratones , Nanopartículas/química , Nanopartículas/uso terapéutico , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/terapia , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/terapia , Sulfuros/química , Sulfuros/farmacología
19.
Acta Biomater ; 74: 334-343, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29753138

RESUMEN

Ternary copper-based chalcogenide nanomaterials have become rather attractive due to the near-infrared (NIR) response in cancer theranostic fields. However, it is still challenging to further improve the theranostic efficiency of these nanomaterials. Herein, Cu-Sb-S nanoparticles (NPs) around 24 nm are synthesized facilely and functionalized with poly(vinylpyrrolidone) (PVP). Under the NIR irradiation, the resultant PVP-Cu-Sb-S NPs exhibit a relatively high photothermal conversion efficiency of 53.16% and a simultaneous reactive oxygen species (ROS) generation effect. Due to these outstanding photothermal/photodynamic effects, excellent tumor ablation results can be achieved by the combination of PVP-Cu-Sb-S NPs and 808 nm NIR laser treatments without obvious side effect. In addition, they show remarkable contrast enhancement according to in vitro and in vivo photoacoustic (PA) imaging. These PVP-Cu-Sb-S NPs could be served as a multifunctional nanotheranostic agent for PA imaging, photothermal/photodynamic cancer therapy. STATEMENT OF SIGNIFICANCE: Highly theranostic efficiency ternary copper-based chalcogenide nanomaterials has not been fully developed yet. Herein we report the PVP-Cu-Sb-S nanoparticles (NPs) with relatively high photothermal efficiency, simultaneous reactive oxygen species generation effect and photoacoustic imaging capability. The photothermal conversion efficiency of PVP-Cu-Sb-S NPs is higher than most of copper-based chalcogenide nanomaterials reported before. These findings provide a new kind of ternary copper-based chalcogenide with an enhanced theranostic effect, which could be served as a promising multifunctional nanotheranostic agent in the field of biomedical application.


Asunto(s)
Hipertermia Inducida/métodos , Rayos Infrarrojos , Neoplasias Experimentales , Técnicas Fotoacústicas/métodos , Fotoquimioterapia/métodos , Animales , Antimonio/química , Antimonio/farmacología , Cobre/química , Cobre/farmacología , Femenino , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Nanopartículas/uso terapéutico , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Povidona/química , Povidona/farmacología , Sulfuros/química , Sulfuros/farmacología , Nanomedicina Teranóstica/métodos
20.
Oncotarget ; 9(4): 4897-4914, 2018 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-29435150

RESUMEN

OBJECTIVE: To investigate the effects of the microbubble (MB) dose, mechanism index (MI) and sonication duration on blood-brain barrier (BBB) disruption induced by diagnostic ultrasound combined with MBs as well as to investigate the potential molecular mechanism. RESULTS: The extent of BBB disruption increased with MB dose, MI and sonication duration. A relatively larger extent of BBB disruption associated with minimal tissue damage was achieved by an appropriate MB dose and ultrasound exposure parameters with diagnostic ultrasound. Decreased expression of ZO-1, occludin and claudin-5 were correlated with disruption of the BBB, as confirmed by paracellular passage of the tracer lanthanum nitrate into the brain parenchyma after BBB disruption. CONCLUSIONS: These findings indicated that this technique is a promising tool for promoting brain delivery of diagnostic and therapeutic agents in the diagnosis and treatment of brain diseases. METHODS: The extent of BBB disruption was qualitatively assessed by Evans blue (EB) staining and quantitatively analyzed by an EB extravasation measurement. A histological examination was performed to evaluate tissue damage. Expression of tight junction (TJ) related proteins ZO-1, occludin and claudin-5 was determined by western blotting analysis and immunohistofluorescence. Transmission electron microscopy was performed to observe ultrastructure changes of TJs after BBB disruption.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA