Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Acta Biomater ; 2024 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-39278301

RESUMO

The regulation of intracellular ionic homeostasis to trigger antigen-specific immune responses has attracted extensive interest in tumor therapy. In this study, we developed a dual-pathway nanoreactor, Au-Cu2-xSe@ZIF-8@P18 NPs (ACS-Z-P NPs), which targets danger-associated molecular patterns (DAMPs) and releases Zn2+ and reactive oxygen species (ROS) within the tumor microenvironment (TME). Zn2+ released from the metal-organic frameworks (MOFs) was deposited in the cytoplasm, leading to aberrant transcription levels of intracellular zinc-regulated proteins and DNA damage, thereby inducing pyroptosis and immunogenic cell death (ICD) dependent on caspase1/gasdermin D (GSDMD) pathway. Furthermore, upon laser irradiation, ACS-Z-P NPs could break through the limitations of inherent defects of immunosuppression in TME, enhance ROS generation through a Fenton-like reaction cascade, which subsequently triggered the activation of inflammatory vesicles and the release of damage-associated molecular patterns (DAMPs). This cascade effect led to the amplification of pyroptosis and immunogenic cell death (ICD), thereby remodeling the immunosuppressed TME. Consequently, this process improved dendritic cell (DC) antigen presentation and augmented anti-tumor T-cell responses, effectively initiating antigen-specific immune responses and further enhancing pyroptosis and ICD. This study explores the therapeutic properties of these mechanisms in detail. STATEMENT OF SIGNIFICANCE: The synthesized Au-Cu2-xSe@ZIF-8@P18 nanoparticles (ACS-Z-Ps) can effectively enhance the body's immune response by regulating zinc ion levels within cells. This regulation leads to abnormal levels of zinc-regulated protein transcription and DNA damage, which induces cellular pyroptosis. As a result, antigen presentation to dendritic cells (DCs) is improved, and anti-tumor T-cell responses are enhanced. The ACS-Z-P NPs overcome the limitations of ROS deficiency and immunosuppression in the tumor microenvironment by using H2O2 in the tumor microenvironment through a Fenton-like reaction. This leads to an increased production of ROS and O2, remodeling of the immunosuppressed tumor microenvironment, and enhanced induction of cell pyroptosis and immunogenic cell death. ACS-Z-P NPs targeted B16 cells using the photosensitizer P18 in combination with PDT treatment. This approach significantly inhibited the proliferation of B16 cells and effectively inhibited tumor growth.

2.
J Nanobiotechnology ; 22(1): 526, 2024 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-39217372

RESUMO

The programmed cell death (PCD) pathway removes functionally insignificant, infection-prone, or potentially tumorigenic cells, underscoring its important role in maintaining the stability of the internal environment and warding off cancer and a host of other diseases. PCD includes various forms, such as apoptosis, copper death, iron death, and cellular pyroptosis. However, emerging solid-state electron-mediated Z-scheme heterostructured semiconductor nanomaterials with high electron-hole (e-h+) separation as a new method for inducing PCD have not been well studied. We synthesize the Bi2S3-Bi2O3-Au-PEG nanorods (BB-A-P NRs) Z-scheme heterostructured semiconductor has a higher redox capacity and biocompatibility. Firstly, the BB-A-P NRs are excited by near-infrared (NIR) light, which mimics the action of catalase by supplying oxygen (O2) and converting it to a single-linear state of oxygen (1O2) via e-h+ transfer. Secondly, they react with hydrogen peroxide (H2O2) and water (H2O) in tumor to produce hydroxyl radicals (•OH), inducing apoptosis. Intriguingly, the Caspase-1/Gasdermin D (GSDMD)-dependent conventional pyroptosis pathway induced cellular pyroptosis activated by apoptosis and reactive oxygen species (ROS) which causes the intense release of damage associated molecular patterns (DAMPs), leading to the inflammatory death of tumor cells. This, in turn, activates the immunological environment to achieve immunogenic cell death (ICD). BB-A-P enables computed tomography imaging, which allows for visualization of the treatment. BB-A-P activated dual PCD can be viewed as an effective mode of cell death that coordinates the intracellular environment, and the various pathways are interrelated and mutually reinforcing which shows promising therapeutic effects and provides a new strategy for eliminating anoxic tumors.


Assuntos
Apoptose , Semicondutores , Animais , Apoptose/efeitos dos fármacos , Camundongos , Linhagem Celular Tumoral , Elétrons , Humanos , Melanoma/patologia , Nanotubos/química , Nanoestruturas/química , Espécies Reativas de Oxigênio/metabolismo , Peróxido de Hidrogênio , Bismuto/química , Piroptose/efeitos dos fármacos , Ouro/química
3.
Analyst ; 149(19): 4889-4898, 2024 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-39171410

RESUMO

Nanozymes, serving as synthetic alternatives to natural enzymes, offer several benefits including cost-effectiveness, enzyme-like catalytic abilities, enhanced stability, adjustable catalytic activity, easy recyclability, mild reaction conditions, and environmental friendliness. Nonetheless, the ongoing quest to develop nanozymes with enhanced activity and to delve into the catalytic mechanism remains a challenge. In our research, we effectively developed Au@CuO nanocomposites (Au@CuO Nc), replicating the functions of four enzymes found in nature: peroxidase (POD), catalase (CAT), glutathione peroxidase (GPx), and oxidase (OXD). The catalytic efficiency of Au@CuO Nc for TMB oxidation (oxTMB) was approximately 4.8 times greater than that of plain Cu2O cubes, attributed to the synergistic catalytic impact between the Au element and Cu2O within Au@CuO Nc. Mechanistic studies revealed that the novel Au@CuO Nc nanozyme greatly enhances the decomposition of H2O2 to reactive oxygen species (ROS) intermediates (˙OH, ˙O2- and 1O2), resulting in increased POD-like activity of the single-component Cu2O cubes. When an antioxidant like TA was added to the chromogenic system, it converted oxTMB into a colorless form of TMB, enabling further evaluation of TA. Hence, a colorimetric sensor was developed for the rapid and precise quantitative measurement of TA, demonstrating strong linearity between 0.3 and 2.4 µM and featuring a low detection threshold of 0.25 µM. Moreover, this sensor was effectively utilized for the assessment of TA in actual tea samples. This work innovatively proposes a simplified and reliable strategy for the advanced design of highly effective Cu-based nanozymes, enhancing enzyme-like reactions for simultaneous, on-site colorimetric probing of antioxidants.


Assuntos
Colorimetria , Cobre , Ouro , Taninos , Cobre/química , Ouro/química , Colorimetria/métodos , Taninos/química , Limite de Detecção , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/análise , Catálise , Oxirredução , Nanopartículas Metálicas/química , Materiais Biomiméticos/química , Nanocompostos/química , Benzidinas/química , Catalase/química , Catalase/metabolismo , Polifenóis
4.
Adv Sci (Weinh) ; 11(29): e2400251, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38867396

RESUMO

Photosynthesis, essential for life on earth, sustains diverse processes by providing nutrition in plants and microorganisms. Especially, photosynthesis is increasingly applied in disease treatments, but its efficacy is substantially limited by the well-known low penetration depth of external light. Here, ultrasound-mediated photosynthesis is reported for enhanced sonodynamic tumor therapy using organic sonoafterglow (ultrasound-induced afterglow) nanoparticles combined with cyanobacteria, demonstrating the proof-of-concept sonosynthesis (sonoafterglow-induced photosynthesis) in cancer therapy. Chlorin e6, a typical small-molecule chlorine, is formulated into nanoparticles to stimulate cyanobacteria for sonosynthesis, which serves three roles, i.e., overcoming the tissue-penetration limitations of external light sources, reducing hypoxia, and acting as a sonosensitizer for in vivo tumor suppression. Furthermore, sonosynthetic oxygenation suppresses the expression of hypoxia-inducible factor 1α, leading to reduced stability of downstream SLC7A11 mRNA, which results in glutathione depletion and inactivation of glutathione peroxidase 4, thereby inducing ferroptosis of cancer cells. This study not only broadens the scope of microbial nanomedicine but also offers a distinct direction for sonosynthesis.


Assuntos
Cianobactérias , Cianobactérias/metabolismo , Cianobactérias/genética , Camundongos , Animais , Humanos , Fotossíntese , Nanopartículas , Neoplasias/terapia , Neoplasias/metabolismo , Porfirinas/metabolismo , Modelos Animais de Doenças , Linhagem Celular Tumoral , Clorofilídeos , Ferroptose/genética , Terapia por Ultrassom/métodos
5.
Adv Sci (Weinh) ; 11(36): e2308506, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38943265

RESUMO

Collaboration between cancer treatment and inflammation management has emerged as an integral facet of comprehensive cancer care. Nevertheless, the development of interventions concurrently targeting both inflammation and cancer has encountered significant challenges stemming from various external factors. Herein, a bioactive agent synthesized by genetically engineering melanin-producing Bacillus thuringiensis (B. thuringiensis) bacteria, simultaneously achieves eco-friendly photothermal agent and efficient reactive oxygen/nitrogen species (RONS) scavenger benefits, perfectly tackling present toughies from inflammation to cancer therapies. The biologically derived melanin exhibits exceptional photothermal-conversion performance, facilitating potent photonic hyperthermia that effectively eradicates tumor cells and tissues, thereby impeding tumor growth. Additionally, the RONS-scavenging properties of melanin produced by B. thuringiensis bacteria contribute to inflammation reduction, augmenting the efficacy of photothermal tumor repression. This study presents a representative paradigm of genetic engineering in B. thuringiensis bacteria to produce functional agents tailored for diverse biomedical applications, encompassing inflammation and cancer therapy.


Assuntos
Bacillus thuringiensis , Engenharia Genética , Melaninas , Bacillus thuringiensis/genética , Bacillus thuringiensis/metabolismo , Melaninas/metabolismo , Melaninas/biossíntese , Camundongos , Animais , Engenharia Genética/métodos , Anti-Inflamatórios/metabolismo , Antineoplásicos/farmacologia , Modelos Animais de Doenças , Inflamação/metabolismo , Inflamação/genética , Humanos , Linhagem Celular Tumoral
6.
Eur Arch Otorhinolaryngol ; 281(8): 3929-3941, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38625559

RESUMO

PURPOSE: To evaluate literature evidences about the efficacy and safety of anti-angiogenesis agents plus chemoradiotherapy versus chemoradiotherapy in the treatment of locally advanced nasopharyngeal carcinoma. METHODS: The relevant literature was systematically searched from the date of establishment to April 2023 in PubMed, Embase, Web of Science, The Cochrane Library, Chinese National Knowledge Infrastructure, Chinese Biological Medicine, Wanfang and VIP database. Search terms included: Nasopharyngeal Neoplasms, Angiogenesis inhibitors, Endostar, Anlotinib, Apatinib, Bevacizumab, Sunitinib, Pazopanib, Chemoradiotherapy. The literature was strictly screened according to the inclusion and exclusion criteria, and 8 eligible studies were finally included in our meta-analysis (4 randomized controlled trials and 4 retrospective studies). RESULTS: A total of 642 patients were included, with 316 in the anti-angiogenesis agents plus chemoradiotherapy group and 326 in the chemoradiotherapy group. The results of our meta-analysis showed that compared with chemoradiotherapy group, the complete response rate (RR = 1.35, 95% CI 1.05-1.74, P = 0.02), objective response rate (RR = 1.26, 95% CI 1.12-1.43, P = 0.0002) in the anti-angiogenesis agents plus chemoradiotherapy group were significantly improved. In terms of safety, there was a higher incidence of cardiac arrhythmia (RR = 3.63, 95% CI 1.16-11.37, P = 0.03) and hypertension (RR = 1.85, 95% CI 1.04-3.27, P = 0.004) in the anti-angiogenesis agents plus chemoradiotherapy group, while no statistically significant differences were reported in other adverse reactions (all P > 0.05). CONCLUSION: Compared with chemoradiotherapy, anti-angiogenesis agents plus chemoradiotherapy could bring more benefits in terms of short-term efficacy, particularly by notably improving both complete response rate and objective response rate, and overall adverse reactions were acceptable. Anti-angiogenesis agents plus chemoradiotherapy may provide a promising direction for the treatment of locally advanced nasopharyngeal carcinoma. SYSTEMATIC REVIEW REGISTRATION: https://inplasy.com/inplasy-2023-8-0076/ , registration number INPLASY202380076.


Assuntos
Inibidores da Angiogênese , Quimiorradioterapia , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas , Humanos , Inibidores da Angiogênese/uso terapêutico , Neoplasias Nasofaríngeas/terapia , Quimiorradioterapia/métodos , Carcinoma Nasofaríngeo/terapia
7.
Phytomedicine ; 124: 155305, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38176275

RESUMO

BACKGROUND: Hyperuricemia (HUA) is a metabolic disease characterized by a high level of uric acid (UA). The extensive historical application of traditional Chinese medicine (TCM) offers a range of herbs and prescriptions used for the treatment of HUA-related disorders. However, the core herbs in the prescriptions and their mechanisms have not been sufficiently explained. PURPOSE: Our current investigation aimed to estimate the anti-HUA effect and mechanisms of Paeonia veitchii Lynch, an herb with high use frequency identified from data mining of TCM prescriptions. METHODS: Prescriptions for HUA/gout treatment were statistically analyzed through a data mining approach to determine the common nature and use frequency of their composition herbs. The chemical constituents of Paeonia veitchii extract (PVE) were analyzed by UPLC-QTOF-MS/MS, while its UA-lowering effect was further evaluated in adenosine-induced liver cells and potassium oxonate (PO) and hypoxanthine (HX)-induced HUA mice. RESULTS: A total of 225 prescriptions involving 246 herbs were sorted out. The properties, flavors and meridians of the appearing herbs were mainly cold, bitter and liver, respectively, while their efficacy was primarily concentrated on clearing heat and dispelling wind. Further usage frequency analysis yielded the top 20 most commonly used herbs, in which PVE presented significant inhibitory activity (IC50 = 131.33 µg/ml) against xanthine oxidase (XOD), and its constituents showed strong binding with XOD in a molecular docking study and further were experimentally validated through XOD enzymatic inhibition and surface plasmon resonance (SPR). PVE (50 to 200 µg/ml) dose-dependently decreased UA levels by inhibiting XOD expression and activity in BRL 3A liver cells. In HUA mice, oral administration of PVE exhibited a significant UA-lowering effect, which was attributed to the reduction of UA production by inhibiting XOD activity and expression, as well as the enhancement of UA excretion by regulating renal urate transporters (URAT1, GLUT9, OAT1 and ABCG2). Noticeably, all doses of PVE treatment did not cause any liver injury, and displayed a renal protective effect. CONCLUSIONS: Our results first comprehensively clarified the therapeutic effect and mechanisms of PVE against HUA through suppressing UA production and promoting UA excretion with hepatic and renal protection, suggesting that PVE could be a promising UA-lowering candidate with a desirable safety profile for the treatment of HUA and prevention of gout.


Assuntos
Gota , Hiperuricemia , Paeonia , Camundongos , Animais , Hiperuricemia/induzido quimicamente , Ácido Úrico/metabolismo , Xantina Oxidase/metabolismo , Simulação de Acoplamento Molecular , Espectrometria de Massas em Tandem , Rim
8.
Int J Nanomedicine ; 18: 2053-2068, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37101838

RESUMO

Background and Purpose: Luteolin (LUT), a flavonoid found in various plants, has been reported to have potential therapeutic effects in melanoma. However, poor water solubility and low bioactivity have severely restricted the clinical application of LUT. Based on the high reactive oxygen species (ROS) levels in melanoma cells, we developed nanoparticles encapsulating LUT with the ROS-responsive material poly(propylene sulfide)-poly(ethylene glycol) (PPS-PEG) to enhance the water solubility of LUT, accelerate the release of LUT in melanoma cells, and further enhance its anti-melanoma effect, providing a viable solution for the application of LUT nano-delivery systems in melanoma therapy. Methods: In this study, LUT-loaded nanoparticles were prepared with PPS-PEG and named as LUT-PPS-NPs. Dynamic light scattering (DLS) and transmission electron microscopy (TEM) were applied to determine the size and morphology of LUT-PPS-NPs. In vitro studies were carried out to determine the uptake and mechanism of LUT-PPS-NPs by SK-MEL-28 melanoma cells. According to the CCK-8 assay, the cytotoxic effects of LUT-PPS-NPs on human skin fibroblasts (HSF) and SK-MEL-28 cells were assessed. Apoptosis assays, cell migration and invasion assays, and proliferation inhibition assays with low and normal density plating were also applied to test the in vitro anti-melanoma effect. Additionally, melanoma models were established utilizing BALB/c nude mice and initially evaluated the growth inhibitory impact following intratumoral injection of LUT-PPS-NPs. Results: The size of LUT-PPS-NPs was 169.77 ± 7.33 nm with high drug loading (15.05 ± 0.07%). In vitro, cellular assays confirmed that LUT-PPS-NPs were efficiently internalized by SK-MEL-28 cells and showed low cytotoxicity against HSF. Moreover, LUT released from LUT-PPS-NPs significantly inhibited tumor cell proliferation, migration and invasion. Animal experiments showed that LUT-PPS-NPs inhibited tumor growth more than 2-fold compared with the LUT group. Conclusion: In conclusion, the LUT-PPS-NPs developed in our study enhanced the anti-melanoma effect of LUT.


Assuntos
Melanoma , Nanopartículas , Animais , Camundongos , Humanos , Luteolina/farmacologia , Luteolina/uso terapêutico , Camundongos Nus , Espécies Reativas de Oxigênio , Melanoma/tratamento farmacológico , Água , Linhagem Celular Tumoral
9.
Small ; 19(32): e2300976, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37066742

RESUMO

Piezoelectric material-mediated sonodynamic therapy (SDT) has received considerable research interest in cancer therapy. However, the simple applications of conventional piezoelectric materials do not realize the full potential of piezoelectric materials in medicine. Therefore, the energy band structure of a piezoelectric material is modulated in this study to meet the actual requirement for cancer treatment. Herein, an elaborate PEGylated piezoelectric solid solution 0.7BiFeO3 -0.3BaTiO3 nanoparticles (P-BF-BT NPs) is synthesized, and the resultant particles achieve excellent piezoelectric properties and their band structure is tuned via band engineering. The tuned band structure of P-BF-BT NPs is energetically favorable for the synchronous production of superoxide radicals (•O2 - ) and oxygen (O2 ) self-supply via water splitting by the piezoelectric effect. Besides, the P-BF-BT NPs can initiate the Fenton reaction to generate hydroxyl radical (•OH), and thus, chemodynamic therapy (CDT) can be augmented by ultrasound. Detailed in vitro and in vivo research has verified the promising effects of multimodal imaging-guided P-BF-BT NP-mediated synergistic SDT/CDT by the piezo-Fenton process in hypoxic tumor elimination, accompanied by high therapeutic biosafety. The current demonstrates a novel strategy for designing and synthesizing "custom-made" piezoelectric materials for cancer therapy in the future.


Assuntos
Nanopartículas , Neoplasias , Humanos , Engenharia , Radical Hidroxila , Hipóxia , Oxigênio , Linhagem Celular Tumoral , Neoplasias/terapia , Peróxido de Hidrogênio
10.
Int J Biol Macromol ; 236: 124020, 2023 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-36921829

RESUMO

Increasing the formation of reactive oxygen species (ROS) and reducing the elimination of ROS are the two main objectives in the development of novel inorganic sonosensitizers for use in sonodynamic therapy (SDT). Therefore, BTO-Pd-MnO2-HA nanocomplexes with targeted tumor cells and degradable oxygen-producing shells were designed as piezoelectric sonosensitizers for enhancing SDT. The deposition of palladium particles (Pd NPs) leads to the formation of Schottky junctions, promoting the separation of electron-hole pairs and thereby increasing the efficiency of toxic ROS generation in SDT. The tumor microenvironment (TME) triggers the degradation of MnO2, and the released Mn2+ ions catalyze the generation of hydroxyl radicals (•OH) from H2O2 through a Fenton-like reaction. BTO-Pd-MnO2-HA can continuously consume glutathione (GSH) and generate O2, thereby improving the efficiency of SDT and chemodynamic therapy (CDT). A multistep enhanced SDT process mediated by the piezoelectric sonosensitizers BTO-Pd-MnO2-HA was designed, targeted by hyaluronic acid (HA), activated by decomposition in TME, and amplified by deposition of Pd. This procedure not only presents a new alternative for the improvement of sonosensitizers but also widens the application of piezoelectric nanomaterials in biomedicine.


Assuntos
Melanoma , Nanocompostos , Neoplasias , Humanos , Ácido Hialurônico , Peróxido de Hidrogênio , Compostos de Manganês/farmacologia , Espécies Reativas de Oxigênio , Microambiente Tumoral , Óxidos , Melanoma/tratamento farmacológico , Nanocompostos/uso terapêutico , Glutationa , Oxigênio , Linhagem Celular Tumoral
11.
Int J Biol Macromol ; 236: 124003, 2023 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-36907306

RESUMO

Insufficient hydrogen peroxide content in tumor cells, unsuitable pH and low efficiency of commonly used metal catalysts severely affect the efficiency of chemodynamic therapy, resulting in unsatisfactory efficacy of chemodynamic therapy alone. For this purpose, we designed a composite nanoplatform capable of targeting tumors and selectively degrading in the tumor microenvironment (TME) to address these issues. In this work, we synthesized Au@Co3O4 nanozyme inspired by crystal defect engineering. The addition of Au determines the formation of oxygen vacancies, accelerates electron transfer, and enhances redox activity, thus significantly enhancing the superoxide dismutase (SOD)-like and catalase (CAT)-like catalytic activities of the nanozyme. Subsequently, we camouflaged the nanozyme using a biomineralized CaCO3 shell to avoid damage to normal tissues by the nanozyme while effectively encapsulating the photosensitizer IR820, and finally the tumor targeting ability of the nanoplatform was enhanced by the modification of hyaluronic acid. Under near-infrared (NIR) light irradiation, the Au@Co3O4@CaCO3/IR820@HA nanoplatform not only visualizes the treatment with multimodal imaging, but also plays a photothermal sensitizing role through various strategies, while enhancing the enzyme catalytic activity, cobalt ion-mediated chemodynamic therapy (CDT) and IR820-mediated photodynamic therapy (PDT), and achieving the synergistic enhancement of reactive oxygen species (ROS) generation.


Assuntos
Neoplasias , Fotoquimioterapia , Humanos , Espécies Reativas de Oxigênio , Oxigênio , Ácido Hialurônico , Biomineralização , Peróxido de Hidrogênio , Neoplasias/tratamento farmacológico , Linhagem Celular Tumoral , Microambiente Tumoral
12.
Biosens Bioelectron ; 224: 115051, 2023 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-36621084

RESUMO

Accurate quantitative, in situ and temporal tracking imaging of tumor-associated miRNAs in living cells could provide a basis for cancer diagnosis and prognosis. In this strategy, a surface-enhanced Raman scattering (SERS)-fluorescence (FL) dual-spectral sensor (DSS) was constructed based on the nanoscale photophysical properties of AuNPs, mediated by functionalized DNA, to achieve rapid imaging of FL and accurate SERS quantification of intracellular miRNAs. The dual-spectrum sensor in the strategy is highly sensitive, specific and reproducibly stable. The LOD values of the dual spectra were 3.58 pM (SERS) as well as 11.8 pM (FL) with RSD values less than 2.69%. The bispectral sensor self-assembled into a trimer by the lapidation of Y-type DNA under the excitation of the target, generating a stable enhanced electric field coupling; and selected adenine located in the enhanced electric field as the reporter molecule, simplifying the labeling process and variables of the Raman reporter molecule, distinguishing it from other traditional methods. This strategy successfully achieved accurate tracking and quantification of miR-21 in cancer cells and showed good stability in the cells. The reported probes are potential tools for reliable monitoring of biomolecular dynamics in living cells.


Assuntos
Técnicas Biossensoriais , Nanopartículas Metálicas , MicroRNAs , Nanopartículas Metálicas/química , Ouro/química , DNA/química , Análise Espectral Raman/métodos , Fenômenos Eletromagnéticos
13.
Front Oncol ; 12: 994726, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36263226

RESUMO

Objectives: To assess the clinical efficacy and toxicity of nimotuzumab in combination with chemoradiotherapy or chemoradiotherapy alone in the treatment of cervical cancer. Methods: The PubMed, Embase, Cochrane Library, Web of Science, China National Knowledge Infrastructure, China Biomedical Medicine, Wanfang, and VIP databases were systematically searched for relevant literature. Ultimately, six randomised controlled trials (n=393) were included in our meta-analysis. Results: A total of 393 patients were included, of which 197 were in the nimotuzumab combined with chemoradiotherapy group and 196 were in the chemoradiotherapy group. The results of our meta-analysis showed that the complete remission rate (risk ratio [RR] = 1.34, 95% confidence interval [CI]: 1.08-1.65, P = 0.007), objective response rate (RR = 1.30, 95% CI: 1.16-1.44, P < 0.05), and three-year survival rate (RR = 1.27, 95% CI: 1.06-1.51, P = 0.008) in the nimotuzumab combined with chemoradiotherapy group were significantly improved compared with the chemoradiotherapy group. This difference was not statistically significant when comparing the incidence of adverse reactions (such as leukocytopenia, gastrointestinal reaction, radiocystitis, and radioproctitis) between the two groups. Conclusions: Nimotuzumab in combination with chemoradiotherapy has some advantages over chemoradiotherapy alone in the treatment of cervical cancer and does not increase toxicity. Therefore, nimotuzumab has the potential to be an effective treatment for cervical cancer; however, further evidence from large-scale randomised controlled trials is needed.

14.
J Nanobiotechnology ; 20(1): 268, 2022 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-35689218

RESUMO

BACKGROUND: Excessive accumulation of reactive oxygen species (ROS) has been documented as the crucial cellular mechanism of cisplatin-induced ototoxicity. However, numerous antioxidants have failed in clinical studies partly due to inefficient drug delivery to the cochlea. A drug delivery system is an attractive strategy to overcome this drawback. METHODS AND RESULTS: In the present study, we proposed the combination of antioxidant astaxanthin (ATX) and ROS-responsive/consuming nanoparticles (PPS-NP) to combat cisplatin-induced ototoxicity. ATX-PPS-NP were constructed by the self-assembly of an amphiphilic hyperbranched polyphosphoester containing thioketal units, which scavenged ROS and disintegrate to release the encapsulated ATX. The ROS-sensitivity was confirmed by 1H nuclear magnetic resonance spectroscopy, transmission electron microscopy and an H2O2 ON/OFF stimulated model. Enhanced release profiles stimulated by H2O2 were verified in artificial perilymph, the HEI-OC1 cell line and guinea pigs. In addition, ATX-PPS-NP efficiently inhibited cisplatin-induced HEI-OC1 cell cytotoxicity and apoptosis compared with ATX or PPS-NP alone, suggesting an enhanced effect of the combination of the natural active compound ATX and ROS-consuming PPS-NP. Moreover, ATX-PPS-NP attenuated outer hair cell losses in cultured organ of Corti. In guinea pigs, NiRe-PPS-NP verified a quick penetration across the round window membrane and ATX-PPS-NP showed protective effect on spiral ganglion neurons, which further attenuated cisplatin-induced moderate hearing loss. Further studies revealed that the protective mechanisms involved decreasing excessive ROS generation, reducing inflammatory chemokine (interleukin-6) release, increasing antioxidant glutathione expression and inhibiting the mitochondrial apoptotic pathway. CONCLUSIONS: Thus, this ROS-responsive nanoparticle encapsulating ATX has favorable potential in the prevention of cisplatin-induced hearing loss.


Assuntos
Antineoplásicos , Perda Auditiva , Nanopartículas , Ototoxicidade , Animais , Antineoplásicos/uso terapêutico , Antioxidantes/uso terapêutico , Apoptose , Cisplatino/farmacologia , Cobaias , Perda Auditiva/induzido quimicamente , Perda Auditiva/tratamento farmacológico , Perda Auditiva/prevenção & controle , Peróxido de Hidrogênio , Nanopartículas/química , Espécies Reativas de Oxigênio/metabolismo , Xantofilas
15.
Artigo em Inglês | MEDLINE | ID: mdl-34863678

RESUMO

Dammar-20(22)E,24-Diene-3ß,6α,12ß-Triol (YNPT2), as one of the main pharmacological and active components of Panax ginseng, promotes ubiquitination and degradation of hypoxia inducible factor Ia through proteasome, which reduces the content of hypoxia inducible factor Ia in tumor cells. Therefore, it is widely used in tumor inhibition. A sensitive and specific bioanalytical method of liquid chromatography-tandem mass spectrometry (LC-MS/MS) for the quantification of YNPT2 rat plasma has been developed. Buspirone was used as the internal standard (IS). A 50 µl aliquot of rat plasma sample was deproteinized by 150 µl methanol-acetonitrile (1:1,v:v), vortex-mixed for 1 min and centrifuged at 15,000 r/min for 10 min at 4 °C. Then, 120 µl of supernatant was pipetted out into the autosampler vials and analyzed by LC-MS/MS with 10 µl injection volume. Chromatographic separation was performed on an Agilent ZORBAX XDB-C18 column (2.1 × 50 mm, 3.5 µm) with mobile phases consisting of water containing 5 mM ammonium acetate (mobile phase A) and acetonitrile (mobile phase B) at a flow rate of 0.6 ml/min over a total run time of 4.0 min. YNPT2 and buspirone (IS) were detected and quantified using positive electrospray ionization in multiple reaction monitoring (MRM) mode with transitions of m/z 441.4 â†’ 109.1 for YNPT2 and m/z 386.3 â†’ 122.1 for IS. The linear range was 5-2000 ng/ml with the square regression coefficient (r2) of 0.9972, and the lower limit of quantification (LLOQ) was 5 ng/ml. The intra-day and inter-day precision deviations of YNPT2 ranged from 3.8 to 6.9% and 3.5-5.8%, and accuracy error ranged from -7.4-5.9% and -9.2-11.9%. The average extraction recovery of YNPT2 in rat plasma was between the range of 98.5%-102.7%. This method was successfully applied to study the pharmacokinetics of YNPT2 in rats after intragastric administration at a single dose of 10.0 mg/kg and after intravenous injection at a single dose of 2.0 mg/kg.


Assuntos
Cromatografia Líquida/métodos , Ginsenosídeos/sangue , Ginsenosídeos/farmacocinética , Espectrometria de Massas em Tandem/métodos , Animais , Ginsenosídeos/química , Limite de Detecção , Modelos Lineares , Masculino , Panax/química , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes
16.
Toxicol Lett ; 354: 56-64, 2022 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-34757176

RESUMO

The ototoxic mechanisms of cisplatin on the organ of Corti and spiral ganglion neurons have been extensively studied, while few studies have been focused on the stria vascularis (SV). Herein, we verified the functional and morphological impairment in SV induced by a single injection of cisplatin (12 mg/kg, I.P.), represented by a reduction in Endocochlear Potentials (EP) and strial atrophy, and explored underlying mechanisms. Our results revealed increased extravasation of chromatic tracers (Evans blue dye and FITC-dextran) around microvessels after cisplatin exposure. The increased vascular permeability could be attributed to changes of pericytes (PCs) and perivascular-resident macrophage-like melanocytes (PVM/Ms) in number or morphology, as well as the enhanced level of HIF-1α and downstream VEGF. This capillary leakage led to a high accumulation of cisplatin in the perivascular space in SV, and disrupted the integrity of blood-labyrinth barrier (BLB). Also, tight junction (ZO-1) loosening and Na+, K+-ATPase damage was considered to be other critical contributors of BLB breakdown, which resulted in EP drop and consequent hearing loss. This study explored the role of stria vascularis in cisplatin-induced ototoxicity in terms of BLB hyperpermeability and pointed to a novel therapeutic target for the prevention of cisplatin-related hearing loss.


Assuntos
Antineoplásicos/toxicidade , Cisplatino/toxicidade , Cóclea/irrigação sanguínea , Cóclea/efeitos dos fármacos , Ototoxicidade/etiologia , Permeabilidade/efeitos dos fármacos , Estria Vascular/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Masculino , Camundongos
17.
Front Pharmacol ; 11: 999, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32719605

RESUMO

Cisplatin is widely used for the treatment of a number of solid malignant tumors. However, ototoxicity induced by cisplatin is an obstacle to effective treatment of tumors. The basis for this toxicity has not been fully elucidated. It is generally accepted that hearing loss is due to excessive production of reactive oxygen species by cells of the cochlea. In addition, recent data suggest that inflammation may trigger inner ear cell death through endoplasmic reticulum stress, autophagy, and necroptosis, which induce apoptosis. Strategies have been extensively explored by which to prevent, alleviate, and treat cisplatin-induced ototoxicity, which minimize interference with antitumor activity. Of these strategies, none have been approved by the Federal Drug Administration, although several preclinical studies have been promising. This review highlights recent strategies that reduce cisplatin-induced ototoxicity. The focus of this review is to identify candidate agents as novel molecular targets, drug administration routes, delivery systems, and dosage schedules. Animal models of cisplatin ototoxicity are described that have been used to evaluate drug efficacy and side effect prevention. Finally, clinical reports of otoprotection in patients treated with cisplatin are highlighted. For the future, high-quality studies are required to provide reliable data regarding the safety and effectiveness of pharmacological interventions that reduce cisplatin-induced ototoxicity.

18.
Am J Transl Res ; 12(3): 743-757, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32269709

RESUMO

An increased fracture risk is often observed in cancer patients undergoing radiotherapy (RT), particularly at sites within the field of radiation. Therefore, the development of appropriate therapeutic options to prevent RT-induced bone loss is urgently needed. A soluble form of the BMP receptor type 1A fusion protein (mBMPR1A-mFc) serves as an antagonist to endogenous BMPR1A. Previous studies have shown that mBMPR1A-mFc treatment increases bone mass in both ovary-intact and ovariectomized via promoting osteoblastic bone formation and inhibiting osteoclastic bone resorption. The present study was designed to investigate whether mBMPR1A-mFc administration prevents radiation-induced bone deterioration in mice. We constructed an animal model of radiation-induced osteoporosis by exposure to a 2-Gy dose of X-rays. Micro-CT, histomorphometric, bone-turnover, and mechanical analyses showed that mBMPR1A-mFc administration prevented trabecular microarchitecture deterioration after RT because of a marked increase in bone formation and a decrease in bone resorption. Mechanistic studies indicated that mBMPR1A-mFc administration promoted osteoblastogenesis by activating Wnt/Lrp5/ß-catenin signaling while decreasing osteoclastogenesis by inhibiting the RANKL/RANK/OPG pathway. Our novel findings provide solid evidence for the application of mBMPR1A-mFc as a therapeutic treatment for radiation-induced osteoporosis.

19.
J Nanobiotechnology ; 18(1): 53, 2020 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-32192504

RESUMO

BACKGROUND: Ototoxicity is one of the major side effects of platinum-based chemotherapy, especially cisplatin therapy. To date, no FDA approved agents to alleviate or prevent this ototoxicity are available. However, ototoxicity is generally believed to be produced by excessive generation of reactive oxygen species (ROS) in the inner ear, thus leading to the development of various antioxidants, which act as otoprotective agents. Astaxanthin (ATX) is an interesting candidate in the development of new therapies for preventing and treating oxidative stress-related pathologies, owing to its unique antioxidant capacity. METHODS AND RESULTS: In this study, we aimed to evaluate the potential antioxidant properties of ATX in the inner ear by using the HEI-OC1 cell line, zebrafish, and guinea pigs. Because ATX has poor solubility and cannot pass through round window membranes (RWM), we established lipid-polymer hybrid nanoparticles (LPN) for loading ATX. The LPN enabled ATX to penetrate RWM and maintain concentrations in the perilymph in the inner ear for 24 h after a single injection. ATX-LPN were found to have favorable biocompatibility and to strongly affect cisplatin-induced generation of ROS, on the basis of DCFHDA staining in HEI-OC1 cells. JC-1 and MitoTracker Green staining suggested that ATX-LPN successfully reversed the decrease in mitochondrial membrane potential induced by cisplatin in vitro and rescued cells from early stages of apoptosis, as demonstrated by FACS stained with Annexin V-FITC/PI. Moreover, ATX-LPN successfully attenuated OHC losses in cultured organ of Corti and animal models (zebrafish and guinea pigs) in vivo. In investigating the protective mechanism of ATX-LPN, we found that ATX-LPN decreased the expression of pro-apoptotic proteins (caspase 3/9 and cytochrome-c) and increased expression of the anti-apoptotic protein Bcl-2. In addition, the activation of JNK induced by CDDP was up-regulated and then decreased after the administration of ATX-LPN, while P38 stayed unchanged. CONCLUSIONS: To best of our knowledge, this is first study concluded that ATX-LPN as a new therapeutic agent for the prevention of cisplatin-induced ototoxicity.


Assuntos
Nanopartículas/química , Polímeros/farmacologia , Xantofilas/farmacologia , Animais , Antioxidantes , Apoptose/efeitos dos fármacos , Caspase 3 , Linhagem Celular , Cisplatino/farmacologia , Orelha Interna/metabolismo , Cobaias , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Modelos Animais , Polímeros/química , Proteínas Proto-Oncogênicas c-bcl-2 , Espécies Reativas de Oxigênio/metabolismo , Xantofilas/química , Peixe-Zebra
20.
Int J Nanomedicine ; 14: 4211-4227, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31239676

RESUMO

Background: Cisplatin is an extensively used anti-neoplastic agent for the treatment of various solid tumors. However, a high incidence of severe ototoxicity is accompanied by its use in the clinic. Currently, no drugs or therapeutic strategies have been approved for the treatment of cisplatin-induced ototoxicity by the FDA. Purpose: The purpose of this study was to investigate the otoprotective effects of dexamethasone (DEX)-loaded silk-polyethylene hydrogel (DEX-SILK) following round window membrane administration in the cisplatin-induced ototoxicity mouse model. Methods: The morphology, gelation kinetics, viscosity and secondary structure of the DEX-SILK hydrogel were analyzed. DEX concentration in the perilymph was tested at different time points following hydrogel injection on the RWM niche. Cultured cells (HEI-OC1), organ of Corti explants (C57/BL6, P0-2), and cisplatin-induced hearing loss mice model (C57/BL6) were used as in vitro and in vivo models for investigating the otoprotective effects of DEX-SILK hydrogel against cisplatin. Results: Encapsulation of DEX with a loading of 8% (w/v) did not significantly change the silk gelation time, and DEX was evenly distributed in the Silk-PEG hydrogel as visualized by scanning electron microscopy (SEM). The concentration of Silk majorly influenced DEX distribution, morphological characteristics, viscosity, and gelation time. The optimized DEX-SILK hydrogel (8% w/v loading, 15% silk concentration, 10 µl) was administered directly onto the RWM of the guinea pigs. The DEX concentration in the perilymph was maintained above 1 µg/ml for at least 21 days for the DEX-SILK, while it was maintained for less than 6 h in the control sample of free DEX. DEX-SILK (5-60 ng/ml) exhibited significant protective effects against cisplatin-induced cellular ototoxicity and notably reduced the production of reactive oxygen species (ROS). Eventually, pretreatment with DEX-SILK effectively preserved outer hair cells in the cultured organ of Corti explants and demonstrated significant hearing protection at 4, 8, and 16 kHz in the cisplatin-induced hearing loss mice as compared to the effects noted following pretreatment with DEX. Conclusion: These results demonstrated the clinical value of DEX-SILK for the therapy of cisplatin-induced ototoxicity.


Assuntos
Cisplatino/efeitos adversos , Dexametasona/farmacologia , Orelha/patologia , Hidrogéis/química , Injeções , Polietilenoglicóis/química , Seda/química , Animais , Materiais Biocompatíveis/efeitos adversos , Bombyx , Linhagem Celular , Cóclea/efeitos dos fármacos , Cóclea/patologia , Dexametasona/uso terapêutico , Modelos Animais de Doenças , Liberação Controlada de Fármacos , Cobaias , Perda Auditiva/induzido quimicamente , Perda Auditiva/tratamento farmacológico , Camundongos Endogâmicos C57BL , Substâncias Protetoras/farmacologia , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA