Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47.495
Filtrar
1.
Ultrason Sonochem ; 105: 106858, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38564910

RESUMEN

Zinc sulfide/graphitic Carbon Nitride binary nanosheets were synthesized by using a novel sonochemical pathway with high electrocatalytic ability. The as- obtained samples were characterized by various analytical methods such as Transmission Electron Microscopy (TEM), Field emission scanning electron microscopy (FESEM), Energy-dispersive X-ray spectroscopy (EDS), X-ray diffraction analysis (XRD), and X-ray photoelectron spectroscopy (XPS) to evaluate the properties of ZnS@CNS synthesized by this new route. Subsequently, the electrical and electrochemical performance of the proposed electrodes were characterized by using EIS and CV to establish an electroactive ability of the nanocomposites. The complete properties like structural and physical of ZnS@CNS were analyzed. As-prepared binary nanocomposite was applied towards the detection of anticancer drug (flutamide) by various electrochemical methods such as cyclic voltammetry (CV), differential pulse voltammetry (DPV) and amperometry. The glassy carbon electrode modified with a ZnS@CNS composite demonstrates a remarkable electrocatalytic efficiency for detecting flutamide in a pH 7.0 (PBS). The composite modified electrode shows synergistic effect of ZnS and CNS catalyst. The electrochemical sensing performance of the linear range was improved significantly due to high electroactive sites and rapid electron transport pathways. Crucially, the electrochemical method was successfully demonstrated in biological fluids which reveals its potential real-time applicability in the analysis of drug.


Asunto(s)
Antineoplásicos , Electrodos , Grafito , Compuestos de Nitrógeno , Sulfuros , Ondas Ultrasónicas , Compuestos de Zinc , Compuestos de Zinc/química , Sulfuros/química , Antineoplásicos/química , Grafito/química , Flutamida/análisis , Flutamida/química , Técnicas Electroquímicas/métodos , Técnicas de Química Sintética , Electroquímica , Límite de Detección , Catálisis , Nanocompuestos/química , Nanoestructuras/química
2.
Nanoscale ; 16(16): 7976-7987, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38567463

RESUMEN

Collective functionalization of the phytochemicals of medicinal herbs on nanoparticles is emerging as a potential cancer therapeutic strategy. This study presents the facile synthesis of surface-functionalized gold nanoparticles using Bacopa monnieri (Brahmi; Bm) phytochemicals and their therapeutically relevant mechanism of action in the colorectal cancer cell line, HT29. The nanoparticles were characterized using UV-visible spectroscopy, TEM-EDAX, zeta potential analysis, TGA, FTIR and 1H NMR spectroscopy, and HR-LC-MS. The particles (Bm-GNPs) were of polygonal shape and were stable against aggregation. They entered the target cells and inhibited the viability and clonogenicity of the cells with eight times more antiproliferative efficacy (25 ± 1.5 µg mL-1) than Bm extract (Bm-EX). In vitro studies revealed that Bm-GNPs bind tubulin (a protein crucial in cell division and a target of anticancer drugs) and disrupt its helical structure without grossly altering its tertiary conformation. Like other antitubulin agents, Bm-GNPs induced G2/M arrest and ultimately killed the cells, as confirmed using flow cytometry analyses. ZVAD-FMK-mediated global pan-caspase inhibition and the apparent absence of cleaved caspase-3 in treated cells indicated that the death did not involve the classic apoptosis pathway. Cellular ultrastructure analyses, western immunoblots, and in situ immunofluorescence visualization of cellular microtubules revealed microtubule-acetylation-independent induction of autophagy as the facilitator of cell death. Together, the data indicate strong antiproliferative efficacy and a possible mechanism of action for these designer nanoparticles. Bm-GNPs, therefore, merit further investigations, including preclinical evaluations, for their therapeutic potential as inducers of non-apoptotic cell death.


Asunto(s)
Autofagia , Neoplasias Colorrectales , Oro , Nanopartículas del Metal , Humanos , Oro/química , Oro/farmacología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/tratamiento farmacológico , Nanopartículas del Metal/química , Autofagia/efectos de los fármacos , Acetilación , Microtúbulos/metabolismo , Microtúbulos/efectos de los fármacos , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma/tratamiento farmacológico , Células HT29 , Caspasas/metabolismo , Fitoquímicos/farmacología , Fitoquímicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/química , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/química
3.
Nanoscale ; 16(16): 7786-7824, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38568434

RESUMEN

Nanozymes, as a type of nanomaterials with enzymatic catalytic activity, have demonstrated tremendous potential in cancer treatment owing to their unique biomedical properties. However, the heterogeneity of tumors and the complex tumor microenvironment pose significant challenges to the in vivo catalytic efficacy of traditional nanozymes. Drawing inspiration from natural enzymes, scientists are now using biomimetic design to build nanozymes from the ground up. This approach aims to replicate the key characteristics of natural enzymes, including active structures, catalytic processes, and the ability to adapt to the tumor environment. This achieves selective optimization of nanozyme catalytic performance and therapeutic effects. This review takes a deep dive into the use of these biomimetically designed nanozymes in cancer treatment. It explores a range of biomimetic design strategies, from structural and process mimicry to advanced functional biomimicry. A significant focus is on tweaking the nanozyme structures to boost their catalytic performance, integrating them into complex enzyme networks similar to those in biological systems, and adjusting functions like altering tumor metabolism, reshaping the tumor environment, and enhancing drug delivery. The review also covers the applications of specially designed nanozymes in pan-cancer treatment, from catalytic therapy to improved traditional methods like chemotherapy, radiotherapy, and sonodynamic therapy, specifically analyzing the anti-tumor mechanisms of different therapeutic combination systems. Through rational design, these biomimetically designed nanozymes not only deepen the understanding of the regulatory mechanisms of nanozyme structure and performance but also adapt profoundly to tumor physiology, optimizing therapeutic effects and paving new pathways for innovative cancer treatment.


Asunto(s)
Materiales Biomiméticos , Nanoestructuras , Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/terapia , Materiales Biomiméticos/química , Materiales Biomiméticos/uso terapéutico , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Catálisis , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Animales , Microambiente Tumoral/efectos de los fármacos , Biomimética
4.
J Med Chem ; 67(8): 6673-6686, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38569098

RESUMEN

The use of benzimidazole-based trinuclear ruthenium(II)-arene complexes (1-3) to selectively target the rare cancer rhabdomyosarcoma is reported. Preliminary cytotoxic evaluations of the ruthenium complexes in an eight-cancer cell line panel revealed enhanced, selective cytotoxicity toward rhabdomyosarcoma cells (RMS). The trinuclear complex 1 was noted to show superior short- and long-term cytotoxicity in RMS cell lines and enhanced selectivity relative to cisplatin. Remarkably, 1 inhibits the migration of metastatic RMS cells and maintains superior activity in a 3D multicellular spheroid model in comparison to that of the clinically used cisplatin. Mechanistic insights reveal that 1 effectively induces genomic DNA damage, initiates autophagy, and prompts the intrinsic and extrinsic apoptotic pathways in RMS cells. To the best of our knowledge, 1 is the first trinuclear ruthenium(II) arene complex to selectively kill RMS cells in 2D and 3D cell cultures.


Asunto(s)
Antineoplásicos , Apoptosis , Complejos de Coordinación , Rabdomiosarcoma , Rutenio , Humanos , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/patología , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Línea Celular Tumoral , Rutenio/química , Rutenio/farmacología , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/síntesis química , Apoptosis/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Relación Estructura-Actividad , Daño del ADN/efectos de los fármacos , Bencimidazoles/farmacología , Bencimidazoles/química , Bencimidazoles/síntesis química , Autofagia/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos
5.
J Med Chem ; 67(8): 6658-6672, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38569135

RESUMEN

BRD4 is associated with a variety of human diseases, including breast cancer. The crucial roles of amino-terminal bromodomains (BDs) of BRD4 in binding with acetylated histones to regulate oncogene expression make them promising drug targets. However, adverse events impede the development of the BD inhibitors. BRD4 adopts an extraterminal (ET) domain, which recruits proteins to drive oncogene expression. We discovered a peptide inhibitor PiET targeting the ET domain to disrupt BRD4/JMJD6 interaction, a protein complex critical in oncogene expression and breast cancer. The cell-permeable form of PiET, TAT-PiET, and PROTAC-modified TAT-PiET, TAT-PiET-PROTAC, potently inhibits the expression of BRD4/JMJD6 target genes and breast cancer cell growth. Combination therapy with TAT-PiET/TAT-PiET-PROTAC and JQ1, iJMJD6, or Fulvestrant exhibits synergistic effects. TAT-PiET or TAT-PiET-PROTAC treatment overcomes endocrine therapy resistance in ERα-positive breast cancer cells. Taken together, we demonstrated that targeting the ET domain is effective in suppressing breast cancer, providing a therapeutic avenue in the clinic.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Proteínas que Contienen Bromodominio , Proteínas de Ciclo Celular , Proliferación Celular , Factores de Transcripción , Humanos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Femenino , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Animales , Proliferación Celular/efectos de los fármacos , Péptidos/farmacología , Péptidos/química , Línea Celular Tumoral , Ratones , Dominios Proteicos , Ratones Desnudos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo
6.
AAPS PharmSciTech ; 25(5): 91, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664316

RESUMEN

Addressing poor solubility and permeability issues associated with synthetic drugs and naturally occurring active compounds is crucial for improving bioavailability. This review explores the potential of phospholipid complex formulation technology to overcome these challenges. Phospholipids, as endogenous molecules, offer a viable solution, with drugs complexed with phospholipids demonstrating a similar absorption mechanism. The non-toxic and biodegradable nature of the phospholipid complex positions it as an ideal candidate for drug delivery. This article provides a comprehensive exploration of the mechanisms underlying phospholipid complexes. Special emphasis is placed on the solvent evaporation method, with meticulous scrutiny of formulation aspects such as the phospholipid ratio to the drug and solvent. Characterization techniques are employed to understand structural and functional attributes. Highlighting the adaptability of the phospholipid complex, the review discusses the loading of various nanoformulations and emulsion systems. These strategies aim to enhance drug delivery and efficacy in various malignancies, including breast, liver, lung, cervical, and pancreatic cancers. The broader application of the drug phospholipid complex is showcased, emphasizing its adaptability in diverse oncological settings. The review not only explores the mechanisms and formulation aspects of phospholipid complexes but also provides an overview of key clinical studies and patents. These insights contribute to the intellectual and translational advancements in drug phospholipid complexes.


Asunto(s)
Antineoplásicos , Sistemas de Liberación de Medicamentos , Neoplasias , Fosfolípidos , Fosfolípidos/química , Humanos , Sistemas de Liberación de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacocinética , Solubilidad , Animales , Química Farmacéutica/métodos , Disponibilidad Biológica , Emulsiones/química , Portadores de Fármacos/química , Composición de Medicamentos/métodos
7.
Sci Rep ; 14(1): 9545, 2024 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664493

RESUMEN

An essential research area for scientists is the development of high-performing, inexpensive, non-toxic antibacterial materials that prevent the transfer of bacteria. In this study, pure Bi2WO6 and Bi2WO6/MWCNTs nanocomposite were prepared by hydrothermal method. A series of characterization results by using XRD FTIR, Raman, FESEM, TEM, and EDS analyses, reveal the formation of orthorhombic nanoflakes Bi2WO6 by the addition of NaOH and pH adjustment to 7. Compared to pure Bi2WO6, the Bi2WO6/MWCNTs nanocomposite exhibited that CNTs are efficiently embedded into the structure of Bi2WO6 which results in charge transfer between metal ion electrons and the conduction or valence band of Bi2WO6 and MWCNTs and result in shifting to longer wavelength as shown in UV-visible and PL. The results confirmed that MWCNTs are stuck to the surface of the microflowers, and some of them embedded inside the Bi2WO6 nanoflakes without affecting the structure of Bi2WO6 nanoflakes as demonstrated by TEM. In addition, Pure Bi2WO6 and the Bi2WO6/MWCNTs nanocomposite were tested against P. mirabilis and S. mutans., confirming the effect of addition MWCNTs materials had better antibacterial activity in opposition to both bacterial strains than pure Bi2WO6. Besides, pure Bi2WO6 and the Bi2WO6/MWCNTs nanocomposite tested for cytotoxicity against lung MTT test on Hep-G2 liver cancer cells, and flow-cytometry. Results indicated that pure Bi2WO6 and the Bi2WO6/MWCNTs nanocomposite have significant anti-cancer efficacy against Hep-G2 cells in vitro. In addition, the findings demonstrated that Bi2WO6 and Bi2WO6/MWCNTs triggered cell death via increasing ROS. Based on these findings, it appears that pure Bi2WO6 and the Bi2WO6/MWCNTs nanocomposite have the potential to be developed as nanotherapeutics for the treatment of bacterial infections, and liver cancer.


Asunto(s)
Antibacterianos , Antineoplásicos , Bismuto , Nanocompuestos , Compuestos de Tungsteno , Nanocompuestos/química , Antibacterianos/farmacología , Antibacterianos/química , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Bismuto/química , Bismuto/farmacología , Compuestos de Tungsteno/química , Compuestos de Tungsteno/farmacología , Nanotubos de Carbono/química , Pruebas de Sensibilidad Microbiana , Supervivencia Celular/efectos de los fármacos , Células Hep G2
8.
J Nanobiotechnology ; 22(1): 209, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664830

RESUMEN

BACKGROUND: Vasculogenic mimicry (VM), when microvascular channels are formed by cancer cells independent of endothelial cells, often occurs in deep hypoxic areas of tumors and contributes to the aggressiveness and metastasis of triple-negative breast cancer (TNBC) cells. However, well-developed VM inhibitors exhibit inadequate efficacy due to their low drug utilization rate and limited deep penetration. Thus, a cost-effective VM inhibition strategy needs to be designed for TNBC treatment. RESULTS: Herein, we designed a low-intensity focused ultrasound (LIFU) and matrix metalloproteinase-2 (MMP-2) dual-responsive nanoplatform termed PFP@PDM-PEG for the cost-effective and efficient utilization of the drug disulfiram (DSF) as a VM inhibitor. The PFP@PDM-PEG nanodroplets effectively penetrated tumors and exhibited substantial accumulation facilitated by PEG deshielding in a LIFU-mediated and MMP-2-sensitive manner. Furthermore, upon exposure to LIFU irradiation, DSF was released controllably under ultrasound imaging guidance. This secure and controllable dual-response DSF delivery platform reduced VM formation by inhibiting COL1/pro-MMP-2 activity, thereby significantly inhibiting tumor progression and metastasis. CONCLUSIONS: Considering the safety of the raw materials, controlled treatment process, and reliable repurposing of DSF, this dual-responsive nanoplatform represents a novel and effective VM-based therapeutic strategy for TNBC in clinical settings.


Asunto(s)
Disulfiram , Neoplasias Pulmonares , Metaloproteinasa 2 de la Matriz , Nanopartículas , Neovascularización Patológica , Neoplasias de la Mama Triple Negativas , Disulfiram/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Animales , Femenino , Humanos , Ratones , Línea Celular Tumoral , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Nanopartículas/química , Neovascularización Patológica/tratamiento farmacológico , Ratones Endogámicos BALB C , Ratones Desnudos , Reposicionamiento de Medicamentos , Ondas Ultrasónicas , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/uso terapéutico
9.
J Nat Prod ; 87(4): 984-993, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38587271

RESUMEN

A chemical investigation of the hydrophilic fraction of a cultured Nodularia sp. (NIES-3585) afforded six new cyclic lipopeptides, noducyclamides A1-A4 (1-4) containing 10 amino acid residues and dodecapeptides noducyclamides B1 and B2 (5 and 6). The planar structures of these lipopeptides were elucidated based on the combination of HRMS and 1D and 2D NMR spectroscopic data analyses. These peptides are structurally analogous to laxaphycins and contain the nonproteinogenic amino acids 3-hydroxyvaline and 3-hydroxyleucine and a ß-amino decanoic acid residue. The absolute configurations of the noducyclamides (1-6) were determined by acid hydrolysis, followed by advanced Marfey's analysis. Noducyclamide B1 (5) showed cytotoxic activities against MCF7 breast cancer cell lines with an IC50 value of 3.0 µg/mL (2.2 µM).


Asunto(s)
Cianobacterias , Péptidos Cíclicos , Humanos , Estructura Molecular , Cianobacterias/química , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/química , Lipopéptidos/farmacología , Lipopéptidos/química , Ensayos de Selección de Medicamentos Antitumorales , Células MCF-7 , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Femenino , Resonancia Magnética Nuclear Biomolecular
10.
Cell Rep ; 43(4): 114108, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38615321

RESUMEN

TRP channels are implicated in various diseases, but high structural similarity between them makes selective pharmacological modulation challenging. Here, we study the molecular mechanism underlying specific inhibition of the TRPM7 channel, which is essential for cancer cell proliferation, by the anticancer agent CCT128930 (CCT). Using cryo-EM, functional analysis, and MD simulations, we show that CCT binds to a vanilloid-like (VL) site, stabilizing TRPM7 in the closed non-conducting state. Similar to other allosteric inhibitors of TRPM7, NS8593 and VER155008, binding of CCT is accompanied by displacement of a lipid that resides in the VL site in the apo condition. Moreover, we demonstrate the principal role of several residues in the VL site enabling CCT to inhibit TRPM7 without impacting the homologous TRPM6 channel. Hence, our results uncover the central role of the VL site for the selective interaction of TRPM7 with small molecules that can be explored in future drug design.


Asunto(s)
1-Naftilamina/análogos & derivados , Antineoplásicos , Canales Catiónicos TRPM , Canales Catiónicos TRPM/metabolismo , Canales Catiónicos TRPM/antagonistas & inhibidores , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Células HEK293 , Simulación de Dinámica Molecular , Sitios de Unión , Unión Proteica , Microscopía por Crioelectrón
11.
J Nat Prod ; 87(4): 1230-1234, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38626456

RESUMEN

Three new cyclic heptapeptides, talaromides A-C (1-3), were isolated from cultures produced by the fungus Talaromyces siglerae (Ascomycota), isolated from an unidentified sponge. The structures, featuring an unusual proline-anthranilic moiety, were elucidated by analysis of spectroscopic data and chemical transformations, including the advanced Marfey's method and GITC derivatization. Talaromides A and B inhibited migration activity against PANC-1 human pancreatic cancer cells without significant cytotoxicity.


Asunto(s)
Péptidos Cíclicos , Poríferos , Talaromyces , Talaromyces/química , Animales , Poríferos/microbiología , Humanos , Estructura Molecular , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/química , Péptidos Cíclicos/aislamiento & purificación , Ensayos de Selección de Medicamentos Antitumorales , Biología Marina , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación
12.
J Enzyme Inhib Med Chem ; 39(1): 2343350, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38655602

RESUMEN

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death. FGFR4 has been implicated in HCC progression, making it a promising therapeutic target. We introduce an approach for identifying novel FGFR4 inhibitors by sequentially adding fragments to a common warhead unit. This strategy resulted in the discovery of a potent inhibitor, 4c, with an IC50 of 33 nM and high selectivity among members of the FGFR family. Although further optimisation is required, our approach demonstrated the potential for discovering potent FGFR4 inhibitors for HCC treatment, and provides a useful method for obtaining hit compounds from small fragments.


Asunto(s)
Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Humanos , Relación Estructura-Actividad , Estructura Molecular , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/síntesis química , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo
13.
BMC Microbiol ; 24(1): 140, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38658810

RESUMEN

Kojic acid is a wonderful fungal secondary metabolite that has several applications in the food, medical, and agriculture sectors. Many human diseases become resistant to normal antibiotics and normal treatments. We need to search for alternative treatment sources and understand their mode of action. Aspergillus flavus ASU45 (OL314748) was isolated from the caraway rhizosphere as a non-aflatoxin producer and identified genetically using 18S rRNA gene sequencing. After applying the Box-Behnken statistical design to maximize KA production, the production raised from 39.96 to 81.59 g/l utilizing (g/l) glucose 150, yeast extract 5, KH2PO4 1, MgSO4.7H2O 2, and medium pH 3 with a coefficient (R2) of 98.45%. Extracted KA was characterized using FTIR, XRD, and a scanning electron microscope. Crystalized KA was an effective antibacterial agent against six human pathogenic bacteria (Bacillus cereus, Staphylococcus aureus, Escherichia coli, Klebsiella pneumonia, Serratia marcescens, and Serratia plymuthica). KA achieves high inhibition activity against Bacillus cereus, K. pneumonia, and S. plymuthica at 100 µg/ml concentration by 2.75, 2.85, and 2.85 compared with chloramphenicol which gives inhibition zones 1, 1.1, and 1.6, respectively. Crystalized KA had anticancer activity versus three types of cancer cell lines (Mcf-7, HepG2, and Huh7) and demonstrated high cytotoxic capabilities on HepG-2 cells that propose strong antitumor potent of KA versus hepatocellular carcinoma. The antibacterial and anticancer modes of action were illustrated using the molecular docking technique. Crystalized kojic acid from a biological source represented a promising microbial metabolite that could be utilized as an alternative antibacterial and anticancer agent effectively.


Asunto(s)
Antibacterianos , Antineoplásicos , Aspergillus flavus , Simulación del Acoplamiento Molecular , Pironas , Aspergillus flavus/efectos de los fármacos , Aspergillus flavus/metabolismo , Aspergillus flavus/genética , Pironas/farmacología , Pironas/química , Pironas/metabolismo , Antibacterianos/farmacología , Antibacterianos/química , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Pruebas de Sensibilidad Microbiana , Línea Celular Tumoral , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/metabolismo , Bacterias/clasificación
14.
J Nanobiotechnology ; 22(1): 204, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658948

RESUMEN

As a famous drug delivery system (DDS), mesoporous organosilica nanoparticles (MON) are degraded slowly in vivo and the degraded components are not useful for cell nutrition or cancer theranostics, and superparamagnetic iron oxide nanoparticles (SPION) are not mesoporous with low drug loading content (DLC). To overcome the problems of MON and SPION, we developed mesoporous SPIONs (MSPIONs) with an average diameter of 70 nm and pore size of 3.9 nm. Sorafenib (SFN) and/or brequinar (BQR) were loaded into the mesopores of MSPION, generating SFN@MSPION, BQR@MSPION and SFN/BQR@MSPION with high DLC of 11.5% (SFN), 10.1% (BQR) and 10.0% (SNF + BQR), demonstrating that our MSPION is a generic DDS. SFN/BQR@MSPION can be used for high performance ferroptosis therapy of tumors because: (1) the released Fe2+/3+ in tumor microenvironment (TME) can produce •OH via Fenton reaction; (2) the released SFN in TME can inhibit the cystine/glutamate reverse transporter, decrease the intracellular glutathione (GSH) and GSH peroxidase 4 levels, and thus enhance reactive oxygen species and lipid peroxide levels; (3) the released BQR in TME can further enhance the intracellular oxidative stress via dihydroorotate dehydrogenase inhibition. The ferroptosis therapeutic mechanism, efficacy and biosafety of MSPION-based DDS were verified on tumor cells and tumor-bearing mice.


Asunto(s)
Sistemas de Liberación de Medicamentos , Ferroptosis , Nanopartículas Magnéticas de Óxido de Hierro , Sorafenib , Ferroptosis/efectos de los fármacos , Animales , Nanopartículas Magnéticas de Óxido de Hierro/química , Ratones , Humanos , Sistemas de Liberación de Medicamentos/métodos , Sorafenib/farmacología , Sorafenib/química , Sorafenib/uso terapéutico , Línea Celular Tumoral , Microambiente Tumoral/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Porosidad , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Ratones Endogámicos BALB C
15.
J Nanobiotechnology ; 22(1): 205, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658965

RESUMEN

The elevated level of hydrogen sulfide (H2S) in colon cancer hinders complete cure with a single therapy. However, excessive H2S also offers a treatment target. A multifunctional cascade bioreactor based on the H2S-responsive mesoporous Cu2Cl(OH)3-loaded hypoxic prodrug tirapazamine (TPZ), in which the outer layer was coated with hyaluronic acid (HA) to form TPZ@Cu2Cl(OH)3-HA (TCuH) nanoparticles (NPs), demonstrated a synergistic antitumor effect through combining the H2S-driven cuproptosis and mild photothermal therapy. The HA coating endowed the NPs with targeting delivery to enhance drug accumulation in the tumor tissue. The presence of both the high level of H2S and the near-infrared II (NIR II) irradiation achieved the in situ generation of photothermic agent copper sulfide (Cu9S8) from the TCuH, followed with the release of TPZ. The depletion of H2S stimulated consumption of oxygen, resulting in hypoxic state and mitochondrial reprogramming. The hypoxic state activated prodrug TPZ to activated TPZ (TPZ-ed) for chemotherapy in turn. Furthermore, the exacerbated hypoxia inhibited the synthesis of adenosine triphosphate, decreasing expression of heat shock proteins and subsequently improving the photothermal therapy. The enriched Cu2+ induced not only cuproptosis by promoting lipoacylated dihydrolipoamide S-acetyltransferase (DLAT) heteromerization but also performed chemodynamic therapy though catalyzing H2O2 to produce highly toxic hydroxyl radicals ·OH. Therefore, the nanoparticles TCuH offer a versatile platform to exert copper-related synergistic antitumor therapy.


Asunto(s)
Cobre , Ácido Hialurónico , Sulfuro de Hidrógeno , Mitocondrias , Nanopartículas , Terapia Fototérmica , Profármacos , Tirapazamina , Terapia Fototérmica/métodos , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/farmacología , Animales , Cobre/química , Cobre/farmacología , Ratones , Humanos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Profármacos/farmacología , Profármacos/química , Tirapazamina/farmacología , Tirapazamina/química , Nanopartículas/química , Ácido Hialurónico/química , Línea Celular Tumoral , Neoplasias del Colon/terapia , Neoplasias del Colon/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Ratones Endogámicos BALB C , Antineoplásicos/farmacología , Antineoplásicos/química , Ratones Desnudos
16.
Int J Nanomedicine ; 19: 3589-3605, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38645464

RESUMEN

Purpose: This study aimed to develop a novel and feasible modification strategy to improve the solubility and antitumor activity of resiquimod (R848) by utilizing the supramolecular effect of 2-hydroxypropyl-beta-cyclodextrin (2-HP-ß-CD). Methods: R848-loaded PLGA nanoparticles modified with 2-HP-ß-CD (CD@R848@NPs) were synthesized using an enhanced emulsification solvent-evaporation technique. The nanoparticles were then characterized in vitro by several methods, such as scanning electron microscopy (SEM), differential scanning calorimetry (DSC), Fourier transform infrared (FTIR) spectroscopy, particle size analysis, and zeta potential analysis. Then, the nanoparticles were loaded with IR-780 dye and imaged using an in vivo imaging device to evaluate their biodistribution. Additionally, the antitumor efficacy and underlying mechanism of CD@R848@NPs in combination with an anti-TNFR2 antibody were investigated using an MC-38 colon adenocarcinoma model in vivo. Results: The average size of the CD@R848@NPs was 376 ± 30 nm, and the surface charge was 21 ± 1 mV. Through this design, the targeting ability of 2-HP-ß-CD can be leveraged and R848 is delivered to tumor-supporting M2-like macrophages in an efficient and specific manner. Moreover, we used an anti-TNFR2 antibody to reduce the proportion of Tregs. Compared with plain PLGA nanoparticles or R848, CD@R848@NPs increased penetration in tumor tissues, dramatically reprogrammed M1-like macrophages, removed tumors and prolonged patient survival. Conclusion: The new nanocapsule system is a promising strategy for targeting tumor, reprogramming tumor -associated macrophages, and enhancement immunotherapy.


Asunto(s)
2-Hidroxipropil-beta-Ciclodextrina , Neoplasias del Colon , Imidazoles , Nanopartículas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Macrófagos Asociados a Tumores , Imidazoles/química , Imidazoles/farmacología , Imidazoles/farmacocinética , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Animales , Nanopartículas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , 2-Hidroxipropil-beta-Ciclodextrina/química , Macrófagos Asociados a Tumores/efectos de los fármacos , Línea Celular Tumoral , Ratones , Humanos , Distribución Tisular , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Antineoplásicos/administración & dosificación , Tamaño de la Partícula , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética
17.
Sci Rep ; 14(1): 9223, 2024 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-38649732

RESUMEN

A series of 20 novel gefitinib derivatives incorporating the 1,2,3-triazole moiety were designed and synthesized. The synthesized compounds were evaluated for their potential anticancer activity against EGFR wild-type human non-small cell lung cancer cells (NCI-H1299, A549) and human lung adenocarcinoma cells (NCI-H1437) as non-small cell lung cancer. In comparison to gefitinib, Initial biological assessments revealed that several compounds exhibited potent anti-proliferative activity against these cancer cell lines. Notably, compounds 7a and 7j demonstrated the most pronounced effects, with an IC50 value of 3.94 ± 0.17 µmol L-1 (NCI-H1299), 3.16 ± 0.11 µmol L-1 (A549), and 1.83 ± 0.13 µmol L-1 (NCI-H1437) for 7a, and an IC50 value of 3.84 ± 0.22 µmol L-1 (NCI-H1299), 3.86 ± 0.38 µmol L-1 (A549), and 1.69 ± 0.25 µmol L-1 (NCI-H1437) for 7j. These two compounds could inhibit the colony formation and migration ability of H1299 cells, and induce apoptosis in H1299 cells. Acute toxicity experiments on mice demonstrated that compound 7a exhibited low toxicity in mice. Based on these results, it is proposed that 7a and 7j could potentially be developed as novel drugs for the treatment of lung cancer.


Asunto(s)
Antineoplásicos , Apoptosis , Proliferación Celular , Gefitinib , Neoplasias Pulmonares , Triazoles , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Gefitinib/farmacología , Triazoles/farmacología , Triazoles/química , Triazoles/síntesis química , Apoptosis/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Ratones , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Células A549 , Relación Estructura-Actividad
18.
J Nanobiotechnology ; 22(1): 198, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38649957

RESUMEN

Heat shock protein 90 (HSP90) is overexpressed in numerous cancers, promotes the maturation of numerous oncoproteins and facilitates cancer cell growth. Certain HSP90 inhibitors have entered clinical trials. Although less than satisfactory clinical effects or insurmountable toxicity have compelled these trials to be terminated or postponed, these results of preclinical and clinical studies demonstrated that the prospects of targeting therapeutic strategies involving HSP90 inhibitors deserve enough attention. Nanoparticulate-based drug delivery systems have been generally supposed as one of the most promising formulations especially for targeting strategies. However, so far, no active targeting nano-formulations have succeeded in clinical translation, mainly due to complicated preparation, complex formulations leading to difficult industrialization, incomplete biocompatibility or nontoxicity. In this study, HSP90 and CD44-targeted A6 peptide functionalized biomimetic nanoparticles (A6-NP) was designed and various degrees of A6-modification on nanoparticles were fabricated to evaluate targeting ability and anticancer efficiency. With no excipients, the hydrophobic HSP90 inhibitor G2111 and A6-conjugated human serum albumin could self-assemble into nanoparticles with a uniform particle size of approximately 200 nm, easy fabrication, well biocompatibility and avoidance of hepatotoxicity. Besides, G2111 encapsulated in A6-NP was only released less than 5% in 12 h, which may avoid off-target cell toxicity before entering into cancer cells. A6 peptide modification could significantly enhance uptake within a short time. Moreover, A6-NP continues to exert the broad anticancer spectrum of Hsp90 inhibitors and displays remarkable targeting ability and anticancer efficacy both in hematological malignancies and solid tumors (with colon tumors as the model cancer) both in vitro and in vivo. Overall, A6-NP, as a simple, biomimetic and active dual-targeting (CD44 and HSP90) nanomedicine, displays high potential for clinical translation.


Asunto(s)
Antineoplásicos , Neoplasias del Colon , Proteínas HSP90 de Choque Térmico , Receptores de Hialuranos , Leucemia Mieloide Aguda , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Animales , Línea Celular Tumoral , Ratones , Neoplasias del Colon/tratamiento farmacológico , Antineoplásicos/farmacología , Antineoplásicos/química , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Nanopartículas/química , Sistemas de Liberación de Medicamentos/métodos , Ratones Desnudos , Ratones Endogámicos BALB C , Péptidos/química , Péptidos/farmacología
19.
Mar Drugs ; 22(4)2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38667771

RESUMEN

Algae are used as safe materials to fabricate novel nanoparticles to treat some diseases. Marine brown alga Sargassum vulgare are used to fabricate silver nanoparticles (Sv/Ag-NPs). The characterization of Sv/Ag-NPs was determined by TEM, EDX, Zeta potential, XRD, and UV spectroscopy. The Sv/Ag-NPs were investigated as antioxidant, anticancer, and antibacterial activities against Gram-positive bacteria Bacillus mojavensis PP400982, Staphylococcus caprae PP401704, Staphylococcus capitis PP402689, and Staphylococcus epidermidis PP403851. The activity of the Sv/Ag-NPs was evaluated as hepatoprotective in vitro in comparison with silymarin. The UV-visible spectrum of Sv/Ag-NPs appeared at 442 nm; the size of Sv/Ag-NPs is in range between 6.90 to 16.97 nm, and spherical in shape. Different concentrations of Sv/Ag-NPs possessed antioxidant, anticancer activities against (HepG-2), colon carcinoma (HCT-116), cervical carcinoma (HeLa), and prostate carcinoma (PC-3) with IC50 50.46, 45.84, 78.42, and 100.39 µg/mL, respectively. The Sv/Ag-NPs induced the cell viability of Hep G2 cells and hepatocytes treated with carbon tetrachloride. The Sv/Ag-NPs exhibited antibacterial activities against Staphylococcus caprae PP401704, Staphylococcus capitis PP402689, and Staphylococcus epidermidis PP403851. This study strongly suggests the silver nanoparticles derived from Sargassum vulgare showed potential hepato-protective effect against carbon tetrachloride-induced liver cells, and could be used as anticancer and antibacterial activities.


Asunto(s)
Antibacterianos , Antineoplásicos , Antioxidantes , Nanopartículas del Metal , Sargassum , Plata , Antibacterianos/farmacología , Antibacterianos/química , Humanos , Plata/farmacología , Plata/química , Antioxidantes/farmacología , Antioxidantes/química , Nanopartículas del Metal/química , Sargassum/química , Antineoplásicos/farmacología , Antineoplásicos/química , Línea Celular Tumoral , Pruebas de Sensibilidad Microbiana , Células Hep G2 , Sustancias Protectoras/farmacología , Sustancias Protectoras/química , Staphylococcus epidermidis/efectos de los fármacos , Células HeLa
20.
Mar Drugs ; 22(4)2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38667800

RESUMEN

Two new meroterpenoids, hyrtamide A (1) and hyrfarnediol A (2), along with two known ones, 3-farnesyl-4-hydroxybenzoic acid methyl ester (3) and dictyoceratin C (4), were isolated from a South China Sea sponge Hyrtios sp. Their structures were elucidated by NMR and MS data. Compounds 2-4 exhibited weak cytotoxicity against human colorectal cancer cells (HCT-116), showing IC50 values of 41.6, 45.0, and 37.3 µM, respectively. Furthermore, compounds 3 and 4 significantly suppressed the invasion of HCT-116 cells while also downregulating the expression of vascular endothelial growth factor receptor 1 (VEGFR-1) and vimentin proteins, which are key markers associated with angiogenesis and epithelial-mesenchymal transition (EMT). Our findings suggest that compounds 3 and 4 may exert their anti-invasive effects on tumor cells by inhibiting the expression of VEGFR-1 and impeding the process of EMT.


Asunto(s)
Antineoplásicos , Neoplasias Colorrectales , Transición Epitelial-Mesenquimal , Poríferos , Terpenos , Humanos , Animales , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Poríferos/química , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Terpenos/farmacología , Terpenos/aislamiento & purificación , Terpenos/química , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células HCT116 , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Vimentina/metabolismo , Línea Celular Tumoral , China
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...