Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 534
Filtrar
1.
Appl Environ Microbiol ; 90(4): e0014624, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38557120

RESUMEN

The metal-resistant bacterium Cupriavidus metallidurans occurs in metal-rich environments. In auriferous soils, the bacterium is challenged by a mixture of copper ions and gold complexes, which exert synergistic toxicity. The previously used, self-made Au(III) solution caused a synergistic toxicity of copper and gold that was based on the inhibition of the CupA-mediated efflux of cytoplasmic Cu(I) by Au(I) in this cellular compartment. In this publication, the response of the bacterium to gold and copper was investigated by using a commercially available Au(III) solution instead of the self-made solution. The new solution was five times more toxic than the previously used one. Increased toxicity was accompanied by greater accumulation of gold atoms by the cells. The contribution of copper resistance determinants to the commercially available Au(III) solution and synergistic gold-copper toxicity was studied using single- and multiple-deletion mutants. The commercially available Au(III) solution inhibited periplasmic Cu(I) homeostasis, which is required for the allocation of copper ions to copper-dependent proteins in this compartment. The presence of the gene for the periplasmic Cu(I) and Au(I) oxidase, CopA, decreased the cellular copper and gold content. Transcriptional reporter gene fusions showed that up-regulation of gig, encoding a minor contributor to copper resistance, was strictly glutathione dependent. Glutathione was also required to resist synergistic gold-copper toxicity. The new data indicated a second layer of synergistic copper-gold toxicity caused by the commercial Au(III) solution, inhibition of the periplasmic copper homeostasis in addition to the cytoplasmic one.IMPORTANCEWhen living in auriferous soils, Cupriavidus metallidurans is not only confronted with synergistic toxicity of copper ions and gold complexes but also by different gold species. A previously used gold solution made by using aqua regia resulted in the formation of periplasmic gold nanoparticles, and the cells were protected against gold toxicity by the periplasmic Cu(I) and Au(I) oxidase CopA. To understand the role of different gold species in the environment, another Au(III) solution was commercially acquired. This compound was more toxic due to a higher accumulation of gold atoms by the cells and inhibition of periplasmic Cu(I) homeostasis. Thus, the geo-biochemical conditions might influence Au(III) speciation. The resulting Au(III) species may subsequently interact in different ways with C. metallidurans and its copper homeostasis system in the cytoplasm and periplasm. This study reveals that the geochemical conditions may decide whether bacteria are able to form gold nanoparticles or not.


Asunto(s)
Cupriavidus , Nanopartículas del Metal , Cobre/metabolismo , Oro/toxicidad , Oro/metabolismo , Nanopartículas del Metal/toxicidad , Nanopartículas del Metal/química , Cupriavidus/genética , Cupriavidus/metabolismo , Proteínas Bacterianas/metabolismo , Iones/metabolismo , Suelo , Glutatión/metabolismo , Oxidorreductasas/metabolismo
2.
Int J Mol Sci ; 25(5)2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38474003

RESUMEN

MicroRNA (miRNA) modulation has been identified as a promising strategy for improving the response of human prostate cancer (PCa) to radiotherapy (RT). Studies have shown that mimics or inhibitors of miRNAs could modulate the sensitivity of PCa cells to RT. In addition, pegylated gold nanoparticles have been studied as a therapeutic approach to treat PCa cells and/or vehicles for carrying miRNAs to the inside of cells. Therefore, we evaluated the capacity of hypofractionated RT and pegylated gold nanorods (AuNPr-PEG) to modulate the miRNA signature on PCa cells. Thus, RT-qPCR was used to analyze miRNA-95, miRNA-106-5p, miRNA-145-5p, and miRNA-541-3p on three human metastatic prostate cell lines (PC3, DU145, and LNCaP) and one human prostate epithelial cell line (HprEpiC, a non-tumor cell line) with and without treatment. Our results showed that miRNA expression levels depend on cell type and the treatment combination applied using RT and AuNPr-PEG. In addition, cells pre-treated with AuNPr-PEG and submitted to 2.5 Gy per day for 3 days decreased the expression levels of miRNA-95, miRNA-106, miRNA-145, and miRNA-541-3p. In conclusion, PCa patients submitted to hypofractionated RT could receive personalized treatment based on their metastatic cellular miRNA signature, and AuNPr-PEG could be used to increase metastatic cell radiosensitivity.


Asunto(s)
Nanopartículas del Metal , MicroARNs , Neoplasias de la Próstata , Masculino , Humanos , MicroARNs/genética , Oro/metabolismo , Línea Celular Tumoral , Neoplasias de la Próstata/metabolismo , Polietilenglicoles/metabolismo , Regulación Neoplásica de la Expresión Génica
3.
J Colloid Interface Sci ; 663: 227-237, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38401443

RESUMEN

Outer membrane vesicle-functionalized nanoparticles (OMV-NPs) have attracted significant interest, especially regarding drug delivery applications and vaccines. Here, we report on novel OMV-NPs by applying bioorthogonal click reaction for encapsulating gold nanoparticles (NPs) within outer membrane vesicles (OMVs) by covalent coupling. For this purpose, outer membrane protein A (OmpA), abundant in large numbers (due to 100,000 copies/cell [1]) in OMVs, was modified via the incorporation of the unnatural amino acid p-azidophenylalanine. The azide group was covalently coupled to alkyne-functionalized NPs after incorporation into OmpA. A simplified procedure using low-speed centrifugation (1,000 x g) was developed for preparing OMV-NPs. The OMV-NPs were characterized by zeta potential, Laurdan-based lipid membrane dynamics studies, and the enzymatic activity of functionalized OMVs with surface-displayed nicotinamide adenine dinucleotide oxidase (Nox). In addition, OMVs from attenuated bacteria (ClearColiTM BL21(DE3), E. coli F470) with surface-displayed Nox or antibody fragments were prepared and successfully coupled to AuNPs. Finally, OMV-NPs displaying single-chain variable fragments from a monoclonal antibody directed against epidermal growth factor receptor were applied to demonstrate the feasibility of OMV-NPs for tumor cell targeting.


Asunto(s)
Oro , Nanopartículas del Metal , Oro/metabolismo , Escherichia coli/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo
4.
Pharmacol Rep ; 76(1): 127-139, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38082190

RESUMEN

BACKGROUND: Chronic inflammation in the course of inflammatory bowel disease may result in colon cancer, or colitis-associated colorectal cancer (CACRC). It is well established that CACRC is associated with oxidative stress and secretion of multiple pro-inflammatory cytokines, e.g. tumor necrosis factor-α. Recently, we proved that the administration of gold(III) complexes resulted in the alleviation of acute colitis in mice. The aim of the current study was to assess the antitumor effect of a novel series of gold(III) complexes: TGS 121, 404, 512, 701, 702, and 703. MATERIALS: Analyzed gold(III) complexes were screened in the in vitro studies using colorectal cancer and normal colon epithelium cell lines, SW480, HT-29, and CCD 841 CoN, and in vivo, in the CACRC mouse model. RESULTS: Of all tested complexes, TGS 121, 404, and 702 exhibited the strongest anti-tumor effect in in vitro viability assay of colon cancer cell lines and in in vivo CACRC model, in which these complexes decreased the total number of colonic tumors and macroscopic score. We also evidenced that the mechanism of action was linked to the enzymatic antioxidant system and inflammatory cytokines. CONCLUSIONS: TGS 121, 404, and 702 present anti-tumor potential and are an attractive therapeutic option for colorectal cancer.


Asunto(s)
Colitis , Neoplasias del Colon , Neoplasias Colorrectales , Humanos , Ratones , Animales , Oro/farmacología , Oro/metabolismo , Oro/uso terapéutico , Colitis/complicaciones , Colitis/tratamiento farmacológico , Colitis/inducido químicamente , Colon , Neoplasias del Colon/metabolismo , Citocinas/metabolismo , Células HT29 , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Sulfato de Dextran/farmacología , Ratones Endogámicos C57BL
5.
Anal Chem ; 96(2): 926-933, 2024 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-38158373

RESUMEN

Nucleolin (NCL) is a multifunctional nuclear protein that plays significant roles in regulating physiological activities of the cells. However, it remains a challenge to monitor the dynamic distribution and expression of nucleolin within living cells during cell stress processes directly. Here, we designed "turn-on" fluorescent nanoprobes composed of specific AS1411 aptamer and nucleus-targeting peptide on gold nanoparticles (AuNPs) to effectively capture and track the NCL distribution and expression during pyroptosis triggered by electrical stimulation (ES). The distribution of nucleolin in the cell membrane and nucleus can be easily observed by simply changing the particle size of the nanoprobes. The present strategy exhibits obvious advantages such as simple operation, low cost, time saving, and suitability for living cell imaging. The ES can induce cancer cell pyroptosis controllably and selectively, with less harm to the viability of normal cells. The palpable cell nuclear stress responses of cancerous cells, including nucleus wrinkling and nucleolus fusion after ES at 1.0 V were obviously observed. Compared with normal cells (MCF-10A), NCL is overexpressed within cancerous cells (MCF-7 cells) using the as-designed nanoprobes, and the ES can effectively inhibit NCL expression within cancerous cells. The developed NCL sensing platform and ES-based methods hold great potential for cellular studies of cancer-related diseases.


Asunto(s)
Aptámeros de Nucleótidos , Nanopartículas del Metal , Humanos , Nucleolina , Proteínas de Unión al ARN/metabolismo , Oro/metabolismo , Piroptosis , Colorantes , Fosfoproteínas/metabolismo
6.
Acta Biomater ; 175: 293-306, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38159895

RESUMEN

Current antibacterial interventions encounter formidable challenges when confronting intracellular bacteria, attributable to their clustering within phagocytes, particularly macrophages, evading host immunity and resisting antibiotics. Herein, we have developed an intelligent cell membrane-based nanosystem, denoted as MM@DAu NPs, which seamlessly integrates cascade-targeting capabilities with controllable antibacterial functions for the precise elimination of intracellular bacteria. MM@DAu NPs feature a core comprising D-alanine-functionalized gold nanoparticles (DAu NPs) enveloped by a macrophage cell membrane (MM) coating. Upon administration, MM@DAu NPs harness the intrinsic homologous targeting ability of their macrophage membrane to infiltrate bacteria-infected macrophages. Upon internalization within these host cells, exposed DAu NPs from MM@DAu NPs selectively bind to intracellular bacteria through the bacteria-targeting agent, D-alanine present on DAu NPs. This intricate process establishes a cascade mechanism that efficiently targets intracellular bacteria. Upon exposure to near-infrared irradiation, the accumulated DAu NPs surrounding intracellular bacteria induce local hyperthermia, enabling precise clearance of intracellular bacteria. Further validation in animal models infected with the typical intracellular bacteria, Staphylococcus aureus, substantiates the exceptional cascade-targeting efficacy and photothermal antibacterial potential of MM@DAu NPs in vivo. Therefore, this integrated cell membrane-based cascade-targeting photothermal nanosystem offers a promising approach for conquering persistent intracellular infections without drug resistance risks. STATEMENT OF SIGNIFICANCE: Intracellular bacterial infections lead to treatment failures and relapses because intracellular bacteria could cluster within phagocytes, especially macrophages, evading the host immune system and resisting antibiotics. Herein, we have developed an intelligent cell membrane-based nanosystem MM@DAu NPs, which is designed to precisely eliminate intracellular bacteria through a controllable cascade-targeting photothermal antibacterial approach. MM@DAu NPs combine D-alanine-functionalized gold nanoparticles with a macrophage cell membrane coating. Upon administration, MM@DAu NPs harness the homologous targeting ability of macrophage membrane to infiltrate bacteria-infected macrophages. Upon internalization, exposed DAu NPs from MM@DAu NPs selectively bind to intracellular bacteria through the bacteria-targeting agent, enabling precise clearance of intracellular bacteria through local hyperthermia. This integrated cell membrane-based cascade-targeting photothermal nanosystem offers a promising avenue for conquering persistent intracellular infections without drug resistance risks.


Asunto(s)
Infecciones Bacterianas , Nanopartículas del Metal , Nanopartículas , Infecciones Estafilocócicas , Animales , Oro/metabolismo , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Estafilocócicas/tratamiento farmacológico , Membrana Celular , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Macrófagos/metabolismo , Alanina
7.
ACS Appl Mater Interfaces ; 15(50): 58241-58250, 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-38059477

RESUMEN

Gold nanomaterials have been shown to augment radiation therapy both in vitro and in vivo. However, studies on these materials are mostly phenomenological due to nanoparticle heterogeneity and the complexity of biological systems. Even accurate quantification of the particle dose still results in bulk average biases; the effect on individual cells is not measured but rather the effect on the overall population. To perform quantitative nanobiology, we coated glass coverslips uniformly at varying densities with Au nanoparticle preparations with different morphologies (45 nm cages, 25 nm spheres, and 30 nm rods). Consequently, the effect of a specific number of particles per unit area in contact with breast cancer cells growing on the coated surfaces was ascertained. Gold nanocages showed the highest degree of radiosensitization on a per particle basis, followed by gold nanospheres and gold nanorods, respectively. All three materials showed little cytotoxic effect at 0 Gy, but clonogenic survival decreased proportionally with the radiation dose and particle coverage density. A similar trend was seen in vivo in the combined treatment antitumor response in 4T1 tumor-bearing animals. The presence of gold affected the type and quantity of reactive oxygen species generated, specifically superoxide and hydroxyl radicals, and the concentration of nanocages correlated with the development of more numerous double-stranded DNA breaks and increased protein oxidation as measured by carbonylation. This work demonstrates the dependence on morphology and concentration of radiation enhancement by gold nanomaterials and may lead to a novel method to differentiate intra- and extracellular functionalities of gold nanomedicine treatment strategies. It further provides insights that can guide the rational development of gold nanomaterial-based radiosensitizers for clinical use.


Asunto(s)
Nanopartículas del Metal , Nanoestructuras , Fármacos Sensibilizantes a Radiaciones , Animales , Oro/farmacología , Oro/metabolismo , Apoptosis , Fármacos Sensibilizantes a Radiaciones/farmacología
8.
Anal Chem ; 95(49): 18107-18113, 2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-38019640

RESUMEN

Technological advances and methodological innovations in cell signaling pathway analysis will facilitate progress in understanding biological processes, intervening in diseases, and screening drugs. In this work, an elaborate strategy for visualizing and monitoring the transient receptor potential melastatin 7 (TRPM7)-Mg2+ signaling pathway in living cells was constructed through the logical analysis of upstream mRNA and downstream molecules by two individual DNA sensors. The DNA sensors are constructed by modifying the dye-labeled DNA sequences on the surface of gold nanoparticles. By hybridizing with upstream mRNA, Cy5-modified DNA sensor 1 can detect and silence it simultaneously, outputting a red fluorescence signal. When the upstream mRNA is silenced, the concentration of downstream molecules of Mg2+ will be affected and down-regulated. The FAM-modified DNA sensor 2 detects this change and emits a green fluorescence as a signal. Therefore, the dynamic information on TRPM7 mRNA and the Mg2+-mediated signaling pathway can be successfully obtained by fluorescence imaging methods. Furthermore, the TRPM7 mRNA-Mg2+ signaling pathway also affects cell activity and migratory function through cell scratching and other experiments. More importantly, the proposed sensor also shows potential for screening signaling pathway inhibitors. Our work provides a simple and general strategy for the visualization of signaling pathways, which helps to understand the changes in the physiological activities of cancer cells and the causes of carcinogenesis and is crucial for cancer diagnosis and prognosis.


Asunto(s)
Nanopartículas del Metal , Neoplasias , Canales Catiónicos TRPM , Proteínas Serina-Treonina Quinasas/metabolismo , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo , ARN Mensajero/genética , Oro/metabolismo , Transducción de Señal , ADN , Neoplasias/genética
9.
J Control Release ; 363: 235-252, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37739016

RESUMEN

Extracellular vesicles (EVs) are promising therapeutic carriers owing to their ideal size range and intrinsic biocompatibility. However, limited targeting ability has caused major setbacks in the clinical application of EV therapeutics. To overcome this, we genetically engineered natural free streptavidin (SA) on the cellular surface of bone marrow mesenchymal stem cells (BMSCs) and obtained typical EVs from these cells (BMSC-EVs). Biotin-coated gold nanoparticles confirmed the expression of SA on the membrane of EVs, which has a high affinity for biotinylated molecules. Using a squamous cell carcinoma model, we demonstrated that a pH-sensitive fusogenic peptide -modification of BMSC-EVs achieved targetability in the microenvironment of a hypoxic tumor to deliver anti-tumor drugs. Using EGFR+HER2- and EGFR-HER2+ breast cancer models, we demonstrated that anti-EGFR and anti-HER2 modifications of BMSC-EVs were able to specifically deliver drugs to EGFR+ and HER2+ tumors, respectively. Using a collagen-induced arthritis model, we confirmed that anti-IL12/IL23-modified BMSC-EVs specifically accumulated in the arthritic joint and alleviated inflammation. Administration of SA-overexpressing BMSC-EVs has limited immunogenicity and high safety in vivo, suggesting that BMSC-derived EVs are ideal drug delivery vehicle. These representative scenarios of targeting modification suggest that, using different biotinylated molecules, the SA-overexpressing BMSC-EVs could be endowed with different targetabilities, which allows BMSC-EVs to serve as a versatile platform for targeted drug delivery under various situations.


Asunto(s)
Neoplasias de la Mama , Vesículas Extracelulares , Células Madre Mesenquimatosas , Nanopartículas del Metal , MicroARNs , Humanos , Femenino , Oro/metabolismo , Vesículas Extracelulares/metabolismo , Neoplasias de la Mama/metabolismo , Receptores ErbB/metabolismo , MicroARNs/metabolismo , Microambiente Tumoral
10.
Int J Biol Macromol ; 252: 126358, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37598824

RESUMEN

Recurrence and metastasis have been recognized as a great challenge in cancer treatment. Cancer stem cells (CSCs), as a small subset of cancer cells, are closely associated with tumor metastasis and recurrence due to their resistance and multi-differentiation characteristics. Herein, we developed a local injectable hyaluronic acid (HA) composite hydrogel (HAAG) that targets CSCs, which can continuously release all-trans retinoic acid (ATRA) and gold nanoparticles (AuNPs) at tumor sites. The composite hydrogel was endowed with the ability to target CSCs through the specific binding of HA to CD44. ATRA was loaded into HA micelles to induce CSCs to differentiate into normal cancer cells, while AuNPs was incorporated into the hydrogel for photothermal therapy (PTT). HAAG exhibited good injectability, photothermal properties and CSCs targeting ability. HAAG not only significantly inhibited the growth of 4T1 mouse breast cancer cells and 4T1-CSCs in vitro, but also effectively inhibited tumor recurrence and metastasis in a 4T1-CSC mouse model in vivo. Our study provides a novel strategy of local differentiation combined with PTT for inhibiting the recurrence and metastasis of breast cancer.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Animales , Ratones , Ácido Hialurónico/química , Oro/farmacología , Oro/metabolismo , Hidrogeles/farmacología , Línea Celular Tumoral , Recurrencia Local de Neoplasia/patología , Nanopartículas/química , Tretinoina/farmacología , Células Madre Neoplásicas/metabolismo
11.
Clin Immunol ; 253: 109685, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37406980

RESUMEN

Inducing tumor-specific T cell responses and regulating suppressive tumor microenvironments have been a challenge for effective tumor therapy. CpG (ODN), the Toll-like receptor 9 agonist, has been widely used as adjuvants of cancer vaccines to induce T cell responses. We developed a novel adjuvant to improve the targeting of lymph nodes. CpG were modified with lipid and glycopolymers by the combination of photo-induced RAFT polymerization and click chemistry, and the novel adjuvant was termed as lipid-glycoadjuvant@AuNPs (LCpG). OVA protein was used as model antigen and melanoma model was established to test the immunotherapy effect of the adjuvant. In tumor model, the antitumor effect and mechanism of LCpG on the response of CTLs were examined by flow cytometry and cell cytotoxicity assay. The effects of LCpG on macrophage polarization and Tregs differentiation in tumor microenvironment were also studied by cell depletion assay and cytokine neutralization assay. We also tested the therapeutic effect of the combination of the adjuvant and anti-PD-1 treatment. LCpG could be rapidly transported to and retained longer in the lymphoid nodes than unmodified CpG. In melanoma model, LCpG controlled both primary tumor and its metastasis, and established long-term memory. In spleen and tumor draining lymphoid nodes, LCpG activated tumor-specific Tc1 responses, with increased CD8+ T-cell proliferation, antigen-specific Tc1 cytokine production and specific-tumor killing capacity. In tumor microenvironments, antigen-specific Tc1 induced by the LCpG promoted CTL infiltration, skewed tumor associated macrophages to M1 phenotype, regulated Treg and induced proinflammatory cytokines production in a CTL-derived IFN-γ-dependent manner. In vivo cell depletion and adoptive transfer experiments confirmed that antitumor activity of LCpG included vaccine was mainly dependent on CTL-derived IFN-γ. The anti-tumor efficacy of LCpG was dramatically enhanced when combined with anti-PD1 immunotherapy. LCpG was a promising adjuvant for vaccine formulation which could augment tumor-specific Tc1 activity, and regulate tumor microenvironments.


Asunto(s)
Vacunas contra el Cáncer , Melanoma , Nanopartículas del Metal , Animales , Ratones , Microambiente Tumoral , Interferón gamma/metabolismo , Oro/metabolismo , Oro/farmacología , Linfocitos T CD8-positivos , Adyuvantes Inmunológicos , Melanoma/metabolismo , Lípidos/farmacología , Ratones Endogámicos C57BL
12.
Artif Cells Nanomed Biotechnol ; 51(1): 334-345, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37455406

RESUMEN

Metastatic melanoma cancer stem cells are subpopulations linked to tumour development, immunoevasive behaviour, treatment resistance, and metastasis, all of which contribute to a poor prognosis. Photodynamic treatment (PDT) is an alternate strategy to cancer eradication that involves the generation of reactive oxygen species. As a carrier, nanoparticles enable efficient cellular uptake of photosensitizers, improving organelle accumulation and cancer cell targeted therapy. This study considered at the effect of PDT on CD133+ Melanoma Stem Cells utilising an Aluminium Phthalocyanine Gold Nanoparticle (AlPcS4Cl-AuNP) combination. A ligand exchange approach was used to conjugate AlPcS4Cl-PEG-AuNP-COOH and was characterised using UV-Vis, FTIR, DLS and Zeta Potential. Stem cells isolated from the A375 cell line irradiated with a laser at 673.2 nm with a fluency of 5 J/cm2 were evaluated. Furthermore, it was important to study if apoptosis was one of the mechanisms causing to cell death which was substantiated with Annexin V/PI, caspase 3 and p53 analysis. The nanoparticle conjugate mediated PDT promoted apoptotic cell death, showing increased expression of p53 and caspase-3. The study proposed a strategy aimed at extending the understanding of PDT in enhancing the therapy of melanoma, suggesting a probable improved cell death when AlPcS4Cl was conjugated to AuNPs.


Asunto(s)
Melanoma , Nanopartículas del Metal , Fotoquimioterapia , Humanos , Nanoconjugados/uso terapéutico , Oro/metabolismo , Proteína p53 Supresora de Tumor , Línea Celular Tumoral , Melanoma/patología , Fármacos Fotosensibilizantes/farmacología , Apoptosis , Células Madre/metabolismo
13.
ACS Nano ; 17(14): 13377-13392, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37449942

RESUMEN

Intestinal metabolism-related diseases, such as constipation, inflammatory bowel disease, irritable bowel syndrome, and colorectal cancer, could be associated with the dysfunction of intestinal mitochondria. The mitochondria of intestinal epithelial cells are of great significance for promoting intestinal motility and maintaining intestinal metabolism. It is necessary for the prophylaxis and therapy of intestinal metabolism-related diseases to improve mitochondrial function. We investigated the effect of 4,6-diamino-2-pyrimidinethiol-modified gold nanoparticles (D-Au NPs) on intestinal mitochondria and studied the regulatory role of D-Au NPs on mitochondria metabolism-related disease. D-Au NPs improved the antioxidation capability of mitochondria, regulated the mitochondrial metabolism, and maintained intestinal cellular homeostasis via the activation of AMPK and regulation of PGC-1α with its downstream signaling (UCP2 and DRP1), enhancing the intestinal mechanical barrier. D-Au NPs improved the intestinal mitochondrial function to intervene in the emergence of constipation, which could help develop drugs to treat and prevent mitochondrial metabolism-related diseases. Our findings provided an in-depth understanding of the mitochondrial effects of Au NPs for improving human intestinal barriers.


Asunto(s)
Oro , Nanopartículas del Metal , Humanos , Oro/metabolismo , Nanopartículas del Metal/uso terapéutico , Ligandos , Mitocondrias , Estreñimiento/inducido químicamente , Estreñimiento/tratamiento farmacológico , Estreñimiento/metabolismo
14.
Int J Mol Sci ; 24(13)2023 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-37446085

RESUMEN

Cancer remains a leading cause of death globally, and its complexity poses a significant challenge to effective treatment. Cancer stem cells and their markers have become key players in tumor growth and progression. CD133, a marker in various cancer types, is an active research area as a potential therapeutic target. This article explores the role of CD133 in cancer treatment, beginning with an overview of cancer statistics and an explanation of cancer stem cells and their markers. The rise of CD133 is discussed, including its structure, functions, and occurrence in different cancer types. Furthermore, the article covers CD133 as a therapeutic target, focusing on gene therapy, immunotherapy, and approaches to affect CD133 expression. Nanoparticles such as gold nanoparticles and nanoliposomes are also discussed in the context of CD133-targeted therapy. In conclusion, CD133 is a promising therapeutic target for cancer treatment. As research in this area progresses, it is hoped that CD133-targeted therapies will offer new and effective treatment options for cancer patients in the future.


Asunto(s)
Nanopartículas del Metal , Neoplasias , Humanos , Oro/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias/metabolismo , Antígeno AC133/metabolismo , Línea Celular Tumoral
15.
ACS Nano ; 17(12): 11384-11395, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37288703

RESUMEN

Pancreatic cancer, with extremely limited treatment options and poor prognosis, urgently needs a breakthrough in early diagnosis and monitoring. Tumor exosomes (T-Exos) detection is presently one of the most clinically significant liquid biopsy approaches for non-invasive pancreatic cancer early diagnosis, which, unfortunately, cannot be applied as a routine diagnostic tool until a number of obstacles, such as unsatisfactory specificity and sensitivity, as well as labor-intensive purification and analysis procedures by ultracentrifugation and enzyme-linked immunosorbent assay, are overcome. Here, we report a facile nanoliquid biopsy assay for the especially specific, ultrasensitive yet economical T-Exos detection by a dual specific biomarker antigen co-recognition and capturing strategy, which is enabled by grafting two corresponding capture antibodies on magnetic nanoparticles and gold nanoparticles, for the accurate detection of target tumor exosomes. This approach exhibits excellent specificity and ultrahigh sensitivity of detecting as low as 78 pg/mL pancreatic cancer exosome specific protein GPC1. Successful screening of 21 pancreatic cancer samples from 22 normal control cases with the enhanced specificity and sensitivity ensures the promising non-invasive monitoring and diagnosis for early stage pancreatic cancer.


Asunto(s)
Exosomas , Nanopartículas del Metal , Neoplasias Pancreáticas , Humanos , Biomarcadores de Tumor/metabolismo , Exosomas/metabolismo , Oro/metabolismo , Neoplasias Pancreáticas/patología , Detección Precoz del Cáncer , Biopsia , Neoplasias Pancreáticas
16.
Int J Mol Sci ; 24(6)2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36982610

RESUMEN

In the large field of bioactive peptides, peptaibols represent a unique class of compounds. They are membrane-active peptides, produced by fungi of the genus Trichoderma and known to elicit plant defenses. Among the short-length peptaibols, trichogin GA IV is nonhemolytic, proteolysis-resistant, antibacterial, and cytotoxic. Several trichogin analogs are endowed with potent activity against phytopathogens, thus representing a sustainable alternative to copper for plant protection. In this work, we tested the activity of trichogin analogs against a breast cancer cell line and a normal cell line of the same derivation. Lys-containing trichogins showed an IC50 below 12 µM, a peptide concentration not significantly affecting the viability of normal cells. Two analogs were found to be membrane-active but noncytotoxic. They were anchored to gold nanoparticles (GNPs) and further investigated for their ability to act as targeting agents. GNP uptake by cancer cells increased with peptide decoration, while it decreased in the corresponding normal epithelial cells. This work highlights the promising biological properties of peptaibol analogs in the field of cancer therapy either as cytotoxic molecules or as active targeting agents in drug delivery.


Asunto(s)
Hypocreales , Nanopartículas del Metal , Trichoderma , Oro/farmacología , Oro/metabolismo , Peptaiboles/farmacología , Peptaiboles/metabolismo , Hypocreales/metabolismo , Trichoderma/metabolismo
17.
J Control Release ; 356: 678-690, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36898530

RESUMEN

Macrophages, innate immune cells, are key players in the maintenance of myocardial homeostasis under normal conditions and tissue repair after injury. The infiltration of macrophages into the injured heart makes them a potentially appealing vehicle for noninvasive imaging and targeted drug delivery of myocardial infarction (MI). In this study, we demonstrated the use of surface hydrolysis-designed AuNPs-zwitterionic-glucose to label macrophages and track their infiltration into isoproterenol hydrochloride (ISO)-induced MI sites noninvasively using CT. The AuNPs-zwitterionic-glucose did not affect the viability or cytokine release of macrophages and were highly taken up by these cells. The in vivo CT images were obtained on Day 4, Day 6, Day 7, and Day 9, and the attenuation was seen to increase in the heart over time compared to the Day 4 scan. In vitro analysis also confirmed the presence of macrophages around injured cardiomyocytes. Additionally, we also addressed the concern of cell tracking or merely AuNP tracking, which is the inherent problem for any form of nanoparticle-labeled cell tracking by using zwitterionic and glucose-functionalized AuNPs. The glucose coated on the surface of AuNPs-zwit-glucose will be hydrolyzed in macrophages, forming only zwitterionic protected AuNPs that cannot be taken up again by endogenous cells in vivo. This will greatly improve the accuracy and precision of imaging and target delivery. We believe this is the first study to noninvasively visualize the infiltration of macrophages into MI hearts using CT, which could be used for imaging and evaluating the possibility of macrophage-mediated delivery in infarcted hearts.


Asunto(s)
Nanopartículas del Metal , Infarto del Miocardio , Humanos , Oro/metabolismo , Hidrólisis , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Macrófagos/metabolismo , Miocitos Cardíacos/metabolismo
18.
Chem Commun (Camb) ; 59(21): 3118-3121, 2023 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-36807620

RESUMEN

Hypervalent bispecific gold nanoparticle-anchored aptamer chimeras (AuNP-APTACs) were designed as a new tool of lysosome-targeting chimeras (LYTACs) for efficient degradation of the ATP-binding cassette, subfamily G, isoform 2 protein (ABCG2) to reverse multidrug resistance (MDR) of cancer cells. The AuNP-APTACs could effectively increase the accumulation of drugs in drug-resistant cancer cells and provide comparable efficacy to small-molecule inhibitors. Thus, this new strategy provides a new way to reverse MDR, holding great promise in cancer therapy.


Asunto(s)
Antineoplásicos , Nanopartículas del Metal , Oro/farmacología , Oro/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Resistencia a Antineoplásicos , Línea Celular Tumoral , Resistencia a Múltiples Medicamentos , Antineoplásicos/farmacología
19.
Neurotoxicology ; 95: 12-22, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36623431

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder with progressive memory loss in dementia. Gold nanoparticles (AuNPs) were reported beneficial for human neural stem cells (hNSCs) treated with Amyloid-beta (Aß), but the neuroprotective mechanisms still are unknown. First, the hNSCs induced by Aß to construct AD cell model in vitro and AuNPs was performed to assess the therapeutic effect of Aß-targeted AD treatment. Then, we investigated the effects of AuNPs on hNSCs viability and proinflammatory factors (interleukin 6 and tumor necrosis factor-alpha) by Cell Counting Kit-8 (CCK-8) and enzyme-linked immunosorbent (ELISA). FACS was carried out to determinate Tuj-1 and glial fibrillary acidic protein (GFAP). Reactive oxygen species (ROS) generation and mitochondrial membrane potential was evaluated by ROS and JC-1 assay kit. In addition, miRNA array was used to systematically detect the differential miRNAs. Dual-luciferase reporter assay was applied to verify the targeting relationship between miR-21-5p and the suppressor of cytokine signalling 6(SOCS6). Quantitative PCR (qPCR) and Western blot assessments were also used to detect related gene expression intracellularly or in the supernatant. The results demonstrate that AuNPs co-treatment repressed the high expression of total tau (T-tau), phosphorylated tau (P-tau), and Aß protein, and reduced apoptosis rate of hNSCs. Aß-induced decreased mitochondrial membrane potential and mitochondria in the hNSCs were damaged, while AuNPs co-treatment showed a protective effect on mitochondrial membrane potential. Co-treatment with AuNPs significantly increased dynamin-related protein 1 (DRP1), nuclear respiratory factor 1 (NRF1), and mitochondrial transcription factor A (TFAM) mRNA levels. AuNPs may improve mitochondrial function impairment due to Aß by elevating mitochondrial membrane potential, upregulating regulators of mitochondrial biogenesis, and inhibiting ROS production. hNSCs transfected with miR-21-5p inhibitor reversed AuNPs mediated cytoprotection induced by Aß. AuNPs upregulation of miR-21-5p expression and exert a mitochondrial protective function. Overexpression of miR-21-5p contributes to enhancing the effect of cytoprotection of AuNPs. MiR-21-5p direct targeting SOCS6 and overexpression SOCS6 exerted opposite effects on hNSCs compared with miR-21-5p mimic group. In conclusion, AuNPs can protect hNSCs from Aß injury and decrease mitochondrial damage by regulating the miR-21-5p/SOCS6 pathway.


Asunto(s)
Enfermedad de Alzheimer , Nanopartículas del Metal , MicroARNs , Células-Madre Neurales , Humanos , Péptidos beta-Amiloides/toxicidad , Péptidos beta-Amiloides/metabolismo , Oro/metabolismo , Oro/farmacología , Oro/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Nanopartículas del Metal/toxicidad , Enfermedad de Alzheimer/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/farmacología , Proteínas Supresoras de la Señalización de Citocinas/uso terapéutico
20.
ACS Appl Mater Interfaces ; 15(2): 2679-2692, 2023 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-36598405

RESUMEN

Single vesicle molecular profiling has the potential to transform cancer detection and monitoring by precisely probing cancer-associated extracellular vesicles (EVs) in the presence of normal EVs in body fluids, but it is challenging due to the small EV size, low abundance of antigens on individual vesicles, and a complex biological matrix. Here, we report a facile dual imaging single vesicle technology (DISVT) for surface protein profiling of individual EVs and quantification of target-specific EV subtypes based on direct molecular capture of EVs from diluted biofluids, dual EV-protein fluorescence-light scattering imaging, and fast image analysis using Bash scripts, Python, and ImageJ. Plasmonic gold nanoparticles (AuNPs) were used to label and detect targeted surface protein markers on individual EVs with dark-field light scattering imaging at the single particle level. Monte Carlo calculations estimated that the AuNPs could detect EVs down to 40 nm in diameter. Using the DISVT, we profiled surface protein markers of interest across individual EVs derived from several breast cancer cell lines, which reflected the parental cells. Studies with plasma EVs from healthy donors and breast cancer patients revealed that the DISVT, but not the traditional bulk enzyme-linked immunosorbent assay, detected human epidermal growth factor receptor 2 (HER2)-positive breast cancer at an early stage. The DISVT also precisely differentiated HER2-positive breast cancer from HER2-negative breast cancer. We additionally showed that the amount of tumor-associated EVs was tripled in locally advanced patients compared to that in early-stage patients. These studies suggest that single EV surface protein profiling with DISVT can provide a facile and high-sensitivity method for early cancer detection and quantitative monitoring.


Asunto(s)
Neoplasias de la Mama , Vesículas Extracelulares , Nanopartículas del Metal , Femenino , Humanos , Antígenos , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/metabolismo , Vesículas Extracelulares/metabolismo , Oro/metabolismo , Detección Precoz del Cáncer/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA