Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 681
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 49(8): 4586-4598, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33836082

RESUMEN

The nucleolus is the site of ribosome biosynthesis encompassing the ribosomal DNA (rDNA) locus in a phase separated state within the nucleus. In budding yeast, we find the rDNA locus and Cdc14, a protein phosphatase that co-localizes with the rDNA, behave like a condensate formed by polymer-polymer phase separation, while ribonucleoproteins behave like a condensate formed by liquid-liquid phase separation. The compaction of the rDNA and Cdc14's nucleolar distribution are dependent on the concentration of DNA cross-linkers. In contrast, ribonucleoprotein nucleolar distribution is independent of the concentration of DNA cross-linkers and resembles droplets in vivo upon replacement of the endogenous rDNA locus with high-copy plasmids. When ribosomal RNA is transcribed from the plasmids by Pol II, the rDNA-binding proteins and ribonucleoprotein signals are weakly correlated, but upon repression of transcription, ribonucleoproteins form a single, stable droplet that excludes rDNA-binding proteins from its center. Degradation of RNA-DNA hybrid structures, known as R-loops, by overexpression of RNase H1 results in the physical exclusion of the rDNA locus from the nucleolar center. Thus, the rDNA locus is a polymer-polymer phase separated condensate that relies on transcription and physical contact with RNA transcripts to remain encapsulated within the nucleolus.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Nucléolo Celular/metabolismo , ADN Ribosómico/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Estructuras R-Loop , ARN Polimerasa I/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Nucléolo Celular/genética , Ensayos Clínicos Fase I como Asunto , ADN Ribosómico/genética , Fase G1/efectos de los fármacos , Fase G1/genética , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Hidroliasas/metabolismo , Cinética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Polímeros/química , Polímeros/metabolismo , Proteínas Tirosina Fosfatasas/genética , ARN Polimerasa I/genética , Ribonucleasa H/genética , Ribonucleasa H/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Sirolimus/farmacología , Regulación hacia Arriba , Agua/química , Agua/metabolismo
2.
Nat Chem Biol ; 16(11): 1179-1188, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32989298

RESUMEN

The mitotic kinase AURORA-A is essential for cell cycle progression and is considered a priority cancer target. Although the catalytic activity of AURORA-A is essential for its mitotic function, recent reports indicate an additional non-catalytic function, which is difficult to target by conventional small molecules. We therefore developed a series of chemical degraders (PROTACs) by connecting a clinical kinase inhibitor of AURORA-A to E3 ligase-binding molecules (for example, thalidomide). One degrader induced rapid, durable and highly specific degradation of AURORA-A. In addition, we found that the degrader complex was stabilized by cooperative binding between AURORA-A and CEREBLON. Degrader-mediated AURORA-A depletion caused an S-phase defect, which is not the cell cycle effect observed upon kinase inhibition, supporting an important non-catalytic function of AURORA-A during DNA replication. AURORA-A degradation induced rampant apoptosis in cancer cell lines and thus represents a versatile starting point for developing new therapeutics to counter AURORA-A function in cancer.


Asunto(s)
Antineoplásicos/química , Aurora Quinasa A/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Proteolisis/efectos de los fármacos , Talidomida/química , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis/efectos de los fármacos , Aurora Quinasa A/genética , Benzazepinas/química , Dominio Catalítico , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Replicación del ADN/efectos de los fármacos , Diseño de Fármacos , Femenino , Humanos , Masculino , Terapia Molecular Dirigida , Polietilenglicoles/química , Unión Proteica , Conformación Proteica
3.
J Cell Physiol ; 236(11): 7322-7341, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33934350

RESUMEN

Mesenchymal stem cells (MSCs) have been identified within dental pulp tissues of exfoliated deciduous (SHEDs) and permanent (DPSCs) teeth. Although differences in their proliferative and differentiation properties were revealed, variability in SHEDs and DPSCs responsiveness to growth factors and cytokines have not been studied before. Here, we investigated the influence of interleukin-17 (IL-17) and basic fibroblast growth factor (bFGF) on stemness features of SHEDs and DPSCs by analyzing their proliferation, clonogenicity, cell cycle progression, pluripotency markers expression and differentiation after 7-day treatment. Results indicated that IL-17 and bFGF differently affected SHEDs and DPSCs proliferation and clonogenicity, since bFGF increased proliferative and clonogenic potential of both cell types, while IL-17 similarly affected SHEDs, exerting no effects on adult counterparts DPSCs. In addition, both factors stimulated NANOG, OCT4, and SOX2 pluripotency markers expression in SHEDs and DPSCs showing diverse intracellular expression patterns dependent on MSCs type. As for the differentiation capacity, both factors displayed comparable effects on SHEDs and DPSCs, including stimulatory effect of IL-17 on early osteogenesis in contrast to the strong inhibitory effect showed for bFGF, while having no impact on SHEDs and DPSCs chondrogenesis. Moreover, bFGF combined with IL-17 reduced CD90 and stimulated CD73 expression on both types of MSCs, whereas each factor induced IL-6 expression indicating its' role in IL-17/bFGF-modulated properties of SHEDs and DPSCs. All these data demonstrated that dental pulp MSCs from primary and permanent teeth exert intrinsic features, providing novel evidence on how IL-17 and bFGF affect stem cell properties important for regeneration of dental pulp at different ages.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Pulpa Dental/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Interleucina-17/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Exfoliación Dental , Diente Primario/efectos de los fármacos , Adulto , Células Cultivadas , Niño , Condrogénesis/efectos de los fármacos , Pulpa Dental/citología , Pulpa Dental/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/efectos de los fármacos , Fenotipo , Diente Primario/citología , Diente Primario/metabolismo , Adulto Joven
4.
Biometals ; 34(4): 867-879, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33907910

RESUMEN

Chloro(triethylphosphine)gold(I), (Et3PAuCl hereafter), is an Auranofin (AF)-related compound showing very similar biological and pharmacological properties. Like AF, Et3PAuCl exhibits potent antiproliferative properties in vitro toward a variety of cancer cell lines and is a promising anticancer drug candidate. We wondered whether Et3PAuCl encapsulation might lead to an improved pharmacological profile also considering the likely reduction of unwanted side-reactions that are responsible for adverse effects and for drug inactivation. Et3PAuCl was encapsulated in biocompatible PLGA-PEG nanoparticles (NPs) and the new formulation evaluated in colorectal HCT-116 cancer cells in comparison to the free gold complex. Notably, encapsulated Et3PAuCl (nano-Et3PAuCl hereafter) mostly retains the cellular properties of the free gold complex and elicits even greater cytotoxic effects in colorectal cancer (CRC) cells, mediated by apoptosis and autophagy. Moreover, a remarkable inhibition of two crucial signaling pathways, i.e. ERK and AKT, by nano-Et3PAuCl, was clearly documented. The implications of these findings are discussed.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Nanopartículas/química , Compuestos Orgánicos de Oro/farmacología , Poliésteres/química , Polietilenglicoles/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Cápsulas , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Compuestos Orgánicos de Oro/síntesis química , Compuestos Orgánicos de Oro/química , Células Tumorales Cultivadas
5.
J Nanobiotechnology ; 19(1): 17, 2021 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-33422073

RESUMEN

BACKGROUND: Chemotherapy and gene therapy are used in clinical practice for the treatment of castration-resistant prostate cancer. However, the poor efficiency of drug delivery and serious systemic side effects remain an obstacle to wider application of these drugs. Herein, we report newly designed PEO-PCL micelles that were self-assembled and modified by spermine ligand, DCL ligand and TAT peptide to carry docetaxel and anti-nucleostemin siRNA. RESULTS: The particle size of the micelles was 42 nm, the zeta potential increased from - 12.8 to 15 mV after grafting with spermine, and the optimal N/P ratio was 25:1. Cellular MTT experiments suggested that introduction of the DCL ligand resulted in high toxicity toward PSMA-positive cells and that the TAT peptide enhanced the effect. The expression of nucleostemin was significantly suppressed in vitro and in vivo, and the tumour-inhibition experiment showed that the dual-drug delivery system suppressed CRPC tumour proliferation. CONCLUSIONS: This targeted drug delivery system inhibited the G1/S and G2/M mitotic cycle via synergistic interaction of chemotherapeutics and gene drugs.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Terapia Genética/métodos , Micelas , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular , Docetaxel/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Tamaño de la Partícula , Poliésteres , Neoplasias de la Próstata/patología , ARN Interferente Pequeño/farmacología
6.
J Nanobiotechnology ; 19(1): 18, 2021 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-33422062

RESUMEN

The combination therapy which has been proposed as the strategy for the cancer treatment could achieve a synergistic effect for cancer therapies and reduce the dosage of the applied drugs. On account of the the unique properties as the high absorbed water content, biocompatibility, and flexibility, the targeting nanogels have been considred as a suitable platform. Herein, a non-toxic pH/thermo-responsive hydrogel P(NIPAAm-co-DMAEMA) was synthesized and characterized through the free-radical polymerization and expanded upon an easy process for the preparation of the smart responsive nanogels; that is, the nanogels were used for the efficient and controlled delivery of the anti-cancer drug doxorubicin (DOX) and chemosensitizer curcumin (CUR) simultaneously like a promising strategy for the cancer treatment. The size of the nanogels, which were made, was about 70 nm which is relatively optimal for the enhanced permeability and retention (EPR) effects. The DOX and CUR co-loaded nanocarriers were prepared by the high encapsulation efficiency (EE). It is important to mention that the controlled drug release behavior of the nanocarriers was also investigated. An enhanced ability of DOX and CUR-loaded nanoformulation to induce the cell apoptosis in the HT-29 colon cancer cells which represented the greater antitumor efficacy than the single-drug formulations or free drugs was resulted through the In vitro cytotoxicity. Overall, according to the data, the simultaneous delivery of the dual drugs through the fabricated nanogels could synergistically potentiate the antitumor effects on the colon cancer (CC).


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Curcumina/farmacología , Doxorrubicina/farmacología , Liberación de Fármacos , Nanogeles/química , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Portadores de Fármacos/farmacología , Composición de Medicamentos , Sistemas de Liberación de Medicamentos/métodos , Quimioterapia Combinada , Células HT29 , Humanos , Concentración de Iones de Hidrógeno , Metacrilatos , Nanopartículas , Tamaño de la Partícula
7.
J Nanobiotechnology ; 19(1): 184, 2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34130695

RESUMEN

Gestational trophoblastic tumors seriously endanger child productive needs and the health of women in childbearing age. Nanodrug-based therapy mediated by transporters provides a novel strategy for the treatment of trophoblastic tumors. Focusing on the overexpression of human equilibrative nucleoside transporter 1 (ENT1) on the membrane of choriocarcinoma cells (JEG-3), cytarabine (Cy, a substrate of ENT1)-grafted liposomes (Cy-Lipo) were introduced for the targeted delivery of methotrexate (Cy-Lipo@MTX) for choriocarcinoma therapy in this study. ENT1 has a high affinity for Cy-Lipo and can mediate the endocytosis of the designed nanovehicles into JEG-3 cells. The ENT1 protein maintains its transportation function through circulation and regeneration during endocytosis. Therefore, Cy-Lipo-based formulations showed high tumor accumulation and retention in biodistribution studies. More importantly, the designed DSPE-PEG2k-Cy conjugation exhibited a synergistic therapeutic effect on choriocarcinoma. Finally, Cy-Lipo@MTX exerted an extremely powerful anti-choriocarcinoma effect with fewer side effects. This study suggests that the overexpressed ENT1 on choriocarcinoma cells holds great potential as a high-efficiency target for the rational design of active targeting nanotherapeutics.


Asunto(s)
Citarabina/uso terapéutico , Liposomas/uso terapéutico , Metotrexato/farmacología , Proteínas de Transporte de Nucleósidos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Coriocarcinoma/tratamiento farmacológico , Coriocarcinoma/patología , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Endocitosis , Tranportador Equilibrativo 1 de Nucleósido/química , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Femenino , Células Hep G2 , Humanos , Células MCF-7 , Ratones Endogámicos BALB C , Ratones Desnudos , Tamaño de la Partícula , Ratas Sprague-Dawley , Distribución Tisular
8.
J Mater Sci Mater Med ; 32(3): 23, 2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33675446

RESUMEN

The combination of Fe3O4@Ag superparamagnetic hybrid nanoparticles and nitric oxide (NO) represents an innovative strategy for a localized NO delivery with a simultaneous antibacterial and antitumoral actions. Here, we report the design of Fe3O4@Ag hybrid nanoparticles, coated with a modified and nitrosated chitosan polymer, able to release NO in a biological medium. After their synthesis, physicochemical characterization confirmed the obtention of small NO-functionalized superparamagnetic Fe3O4@Ag NPs. Antibacterial assays demonstrated enhanced effects compared to control. Bacteriostatic effect against Gram-positive strains and bactericidal effect against E. coli were demonstrated. Moreover, NO-functionalized Fe3O4@Ag NPs demonstrated improved ability to reduce cancer cells viability and less cytotoxicity against non-tumoral cells compared to Fe3O4@Ag NPs. These effects were associated to the ability of these NPs act simultaneous as cytotoxic (necrosis inductors) and cytostatic compounds inducing S-phase cell cycle arrest. NPs also demonstrated low hemolysis ratio (<10%) at ideal work range, evidencing their potential for biomedical applications. Targeted and hemocompatible nitric oxide-releasing multi-functional hybrid nanoparticles for antitumor and antimicrobial applications.


Asunto(s)
Compuestos Férricos/química , Nanopartículas del Metal/química , Nanoestructuras/química , Óxido Nítrico/química , Plata/química , Antibacterianos/química , Antibacterianos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Materiales Biocompatibles , Ciclo Celular/efectos de los fármacos , Línea Celular , Humanos , Ensayo de Materiales , Óxido Nítrico/farmacología
9.
Int J Mol Sci ; 22(10)2021 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-34069278

RESUMEN

Clinical outcomes of conventional drug combinations are not ideal due to high toxicity to healthy tissues. Cisplatin (CDDP) is the standard component for many cancer treatments, yet its principal dose-limiting side effect is nephrotoxicity. Thus, CDDP is commonly used in combination with other drugs, such as the autophagy inhibitor chloroquine (CQ), to enhance tumor cell killing efficacy and prevent the development of chemoresistance. In addition, nanocarrier-based drug delivery systems can overcome chemotherapy limitations, decreasing side effects and increasing tumor accumulation. The aim of this study was to evaluate the toxicity of CQ and CDDP against tumor and non-tumor cells when used in a combined treatment. For this purpose, two types of micelles based on Pluronic® F127 hybrid dendritic-linear-dendritic block copolymers (HDLDBCs) modified with polyester or poly(esteramide) dendrons derived from 2,2'-bis(hydroxymethyl)propionic acid (HDLDBC-bMPA) or 2,2'-bis(glycyloxymethyl)propionic acid (HDLDBC-bGMPA) were explored as delivery nanocarriers. Our results indicated that the combined treatment with HDLDBC-bMPA(CQ) or HDLDBC-bGMPA(CQ) and CDDP increased cytotoxicity in tumor cells compared to the single treatment with CDDP. Encapsulations demonstrated less short-term cytotoxicity individually or when used in combination compared to the free drugs. However, and more importantly, a low degree of cytotoxicity against non-tumor cells was maintained, even when drugs were given simultaneously.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Portadores de Fármacos/química , Micelas , Polímeros/química , Células A549 , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cloroquina/administración & dosificación , Cloroquina/farmacocinética , Cisplatino/administración & dosificación , Cisplatino/farmacocinética , Portadores de Fármacos/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Fibroblastos/efectos de los fármacos , Células HeLa , Humanos , Poloxámero/química , Polímeros/síntesis química
10.
Int J Mol Sci ; 22(10)2021 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-34069280

RESUMEN

The use of human dental pulp stromal cells (hDPSCs) has gained increasing attention as an alternative stem cell source for bone tissue engineering. The modification of the cells' epigenetics has been found to play an important role in regulating differentiation, with the inhibition of histone deacetylases 3 (HDAC3) being linked to increased osteogenic differentiation. This study aimed to induce epigenetic reprogramming using the HDAC2 and 3 selective inhibitor, MI192 to promote hDPSCs osteogenic capacity for bone regeneration. MI192 treatment caused a time-dose-dependent change in hDPSC morphology and reduction in viability. Additionally, MI192 successfully augmented hDPSC epigenetic functionality, which resulted in increased histone acetylation and cell cycle arrest at the G2/M phase. MI192 pre-treatment exhibited a dose-dependent effect on hDPSCs alkaline phosphatase activity. Quantitative PCR and In-Cell Western further demonstrated that MI192 pre-treatment significantly upregulated hDPSCs osteoblast-related gene and protein expression (alkaline phosphatase, bone morphogenic protein 2, type I collagen and osteocalcin) during osteogenic differentiation. Importantly, MI192 pre-treatment significantly increased hDPSCs extracellular matrix collagen production and mineralisation. As such, for the first time, our findings show that epigenetic reprogramming with the HDAC2 and 3 selective inhibitor MI192 accelerates the osteogenic differentiation of hDPSCs, demonstrating the considerable utility of this MSCs engineering approach for bone augmentation strategies.


Asunto(s)
Benzamidas/farmacología , Pulpa Dental/citología , Inhibidores de Histona Desacetilasas/farmacología , Isoquinolinas/farmacología , Osteogénesis/efectos de los fármacos , Acetilación/efectos de los fármacos , Fosfatasa Alcalina/metabolismo , Benzamidas/administración & dosificación , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/administración & dosificación , Histonas/metabolismo , Humanos , Isoquinolinas/administración & dosificación , Tercer Molar/citología , Osteogénesis/fisiología , Células del Estroma/metabolismo
11.
Int J Mol Sci ; 22(17)2021 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-34502536

RESUMEN

The advancement of nanotechnology in the last decade has developed an abundance of novel and intriguing TiO2-based nanomaterials that are widely used in many sectors, including industry (as a food additive and colorant in cosmetics, paints, plastics, and toothpaste) and biomedicine (photoelectrochemical biosensing, implant coatings, drug delivery, and new emerging antimicrobial agents). Therefore, the increased use of engineered nanomaterials in the industry has raised serious concern about human exposure and their unexpected cytotoxic effects. Since inhalation is considered the most relevant way of absorbing nanomaterials, different cell death mechanisms induced in MRC-5 lung fibroblasts, following the exposure to functionalized TiO2 NPs, were investigated. Long-term exposure to TiO2 nanoparticles co-doped with 1% of iron and nitrogen led to the alteration of p53 protein activity and the gene expression controlled by this suppressor (NF-kB and mdm2), DNA damage, cell cycle disruptions at the G2/M and S phases, and lysosomal membrane permeabilization and the subsequent release of cathepsin B, triggering the intrinsic pathway of apoptosis in a Bax- and p53-independent manner. Our results are of major significance, contributing to the understanding of the mechanisms underlying the interaction of these nanoparticles with in vitro biological systems, and also providing useful information for the development of new photocatalytic nanoparticles that are active in the visible spectrum, but with increased biocompatibility.


Asunto(s)
Monóxido de Carbono/química , Fibroblastos/efectos de los fármacos , Hierro/química , Nanopartículas del Metal/administración & dosificación , Nitrógeno/química , Titanio/química , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Pulmón/citología , Pulmón/metabolismo , Nanopartículas del Metal/química , Nanopartículas del Metal/ultraestructura , Microscopía Electrónica de Transmisión , Espectroscopía de Fotoelectrones , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/genética , Difracción de Rayos X
12.
Molecules ; 26(11)2021 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-34205043

RESUMEN

Glioblastoma (GBM) is the most common and most deadly primary malignant brain tumor. Current therapies are not effective, the average survival of GBM patients after diagnosis being limited to few months. Therefore, the discovery of new treatments for this highly aggressive brain cancer is urgently needed. Chalcones are synthetic and naturally occurring compounds that have been widely investigated as anticancer agents. In this work, three chalcone derivatives were tested regarding their inhibitory activity and selectivity towards GBM cell lines (human and mouse) and a non-cancerous mouse brain cell line. The chalcone 1 showed the most potent and selective cytotoxic effects in the GBM cell lines, being further investigated regarding its ability to reduce critical hallmark features of GBM and to induce apoptosis and cell cycle arrest. This derivative showed to successfully reduce the invasion and proliferation capacity of tumor cells, both key targets for cancer treatment. Moreover, to overcome potential systemic side effects and its poor water solubility, this compound was encapsulated into liposomes. Therapeutic concentrations were incorporated retaining the potent in vitro growth inhibitory effect of the selected compound. In conclusion, our results demonstrated that this new formulation can be a promising starting point for the discovery of new and more effective drug treatments for GBM.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/metabolismo , Ciclo Celular/efectos de los fármacos , Chalconas/farmacología , Glioblastoma/metabolismo , Animales , Antineoplásicos/química , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Chalconas/química , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Humanos , Liposomas , Ratones , Estructura Molecular , Invasividad Neoplásica
13.
Invest New Drugs ; 38(5): 1236-1246, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31823161

RESUMEN

In this study, we examined the sensitivity of pancreatic cancer cells to [HuArgI (Co)-PEG5000]-induced arginine deprivation as well as the mechanisms underlying deprivation-induced cell death. [HuArgI (Co)-PEG5000]-induced arginine deprivation was cytotoxic to all cell lines tested with IC50 values in the pM range at 72 h post-treatment. Three of the five cell lines were rescued by the addition of excess L-citrulline and expressed ASS1, indicating partial arginine auxotrophy. The remaining two cell lines, on the other hand, were not rescued by the addition of L-citrulline and did not express ASS1, indicating complete auxotrophy to arginine. In addition, all cell lines exhibited G0/G1 cell cycle arrest, in the surviving cell fraction, at 72 h following arginine deprivation. Analysis of the type of cell death revealed negative staining for annexin V and a lack of caspase activation in all five cancer cell lines, following treatment, indicating that arginine deprivation leads to caspase-independent, non-apoptotic cell death. Finally, we demonstrated that arginine deprivation leads to a marked activation of autophagy and that inhibition of this autophagy greatly decreases cytotoxicity, indicating that arginine deprivation induces autophagic cell death in pancreatic cancer cells. We have shown that pancreatic cancer cells are auxotrophic for arginine and sensitive to [HuArgI (Co)-PEG5000]-induced arginine deprivation, hence demonstrating that arginine deprivation is a potentially potent and selective treatment for pancreatic cancer. We have also demonstrated that autophagy is activated following arginine-deprivation and that its prolonged activation leads to autophagic cell death.


Asunto(s)
Antineoplásicos/farmacología , Arginasa/farmacología , Arginina , Muerte Celular Autofágica/efectos de los fármacos , Polietilenglicoles/farmacología , Argininosuccinato Sintasa/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo
14.
Wound Repair Regen ; 28(1): 16-25, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31270882

RESUMEN

Reepithelialization is an important step of wound healing, which is mainly completed by proliferation and migration of epidermal cells. Akermanite is a Ca-, Mg-, and Si-containing bioceramic. This study evaluated the effects of Akermanite on wound healing and investigated the mechanisms. Using scald burn mice models, we demonstrated that local Akermanite treatment significantly accelerated wound healing by increasing reepithelialization and the stemness of epidermal cells. Epidermal cells were cultured in medium containing Akermanite extracts to explore the cellular mechanism of reepithelialization. Akermanite promoted the cell proliferation and migration, maintaining more cells in the S and G2 /M phases of the cell cycle. An additional study showed that Akermanite enhanced the expressions of integrinß1, Lgr4, Lgr5, and Lgr6, which are specific molecular markers of epidermal stem cells, accompanied by the activation of the Wnt/ß-catenin pathway. These results suggested that Akermanite accelerated reepithelialization by increasing the proliferation, migration, and stemness of epidermal cells in a manner related to the Wnt/ß-catenin pathway, which might contribute, at least partially, to accelerated wound healing by Akermanite therapy.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Cerámica/farmacología , Células Epidérmicas/efectos de los fármacos , Repitelización/efectos de los fármacos , Células Madre/metabolismo , Animales , Materiales Biocompatibles , Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Células Epidérmicas/metabolismo , Humanos , Integrina beta1/efectos de los fármacos , Integrina beta1/genética , Integrina beta1/metabolismo , Ratones , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Vía de Señalización Wnt , Cicatrización de Heridas
15.
J Pharmacokinet Pharmacodyn ; 47(2): 163-182, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32162138

RESUMEN

Neutropenia is one of the most common dose-limiting toxocities associated with anticancer drug therapy. The ability to predict the probability and severity of neutropenia based on in vitro studies of drugs in early drug development will aid in advancing safe and efficacious compounds to human testing. Toward this end, a physiological model of granulopoiesis and its regulation is presented that includes the bone marrow progenitor cell cycle, allowing for a mechanistic representation of the action of relevant anticancer drugs based on in vitro studies. Model development used data from previously reported tracer kinetic studies of granulocyte disposition in healthy humans to characterize the dynamics of neutrophil margination in the presence of endogenous granulocyte-colony stimulating factor (G-CSF). In addition, previously published data from healthy volunteers following pegfilgrastim and filgrastim were used to quantify the regulatory effects of support G-CSF therapies on granulopoiesis. The model was evaluated for the cell cycle inhibitor palbociclib, using an in vitro system of human bone marrow mononuclear cells to quantify the action of palbociclib on proliferating progenitor cells, including its inhibitory effect on G1 to S phase transition. The in vitro results were incorporated into the physiological model of granulopoiesis and used to predict the time course of absolute neutrophil count (ANC) and the incidence of neutropenia observed in three previously reported clinical trials of palbociclib. The model was able to predict grade 3 and 4 neutropenia due to palbociclib treatment with 86% accuracy based on in vitro data.


Asunto(s)
Médula Ósea/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Granulocitos/efectos de los fármacos , Neutropenia/inducido químicamente , Algoritmos , Antineoplásicos/farmacología , Movimiento Celular , Filgrastim/farmacología , Factor Estimulante de Colonias de Granulocitos/farmacología , Hematopoyesis/efectos de los fármacos , Humanos , Recuento de Leucocitos , Modelos Biológicos , Neutrófilos/efectos de los fármacos , Piperazinas/farmacología , Polietilenglicoles/farmacología , Piridinas/farmacología , Células Madre/efectos de los fármacos
16.
Chem Biodivers ; 17(9): e2000369, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32644295

RESUMEN

The current study was carried out by a bioguided fractionation of a hexane extract of the latex of Euphorbia umbellata against leukemic cells. Samples were analyzed by NMR, GC/MS, triterpenes quantification, and MTT reduction assay. Morphological, cell cycle, mitochondrial membrane potential and caspases 3/7 analyses were performed for the dichloromethane and ethanol fractions, and selectivity index for the dichloromethane fraction. NMR analysis presented characteristic signals of terpenes and steroids, data were confirmed by the quantification of triterpenes and GC/MS analysis. MTT reduction assay demonstrated that HL-60 was the most sensitive cell lineage against dichloromethane and ethanol fractions. Compounds of these matrices caused morphological changes compatible with apoptosis induction, altered cell cycle, increment of depolarized population cells and activation of caspases 3/7. Selectivity indices were higher than 22.44. Bioguided-fractionation study showed that samples of the latex of E. umbellata raised the activity of the phytocomplex against leukemic cells, and the cytotoxicity can be associated with an apoptosis pathway.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Euphorbia/química , Látex/química , Terpenos/farmacología , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Terpenos/química , Terpenos/aislamiento & purificación , Células Tumorales Cultivadas
17.
J Cell Physiol ; 234(12): 22285-22298, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31073992

RESUMEN

Silibinin, a polyphenolic flavonolignan, is well-known as a safe therapeutic drug without any side effects in the treatment of many malignancies especially cancerous cells. In this study, to overcome problems such as low solubility of silibinin and to enhance its delivery to cancerous cells, we encapsulated silibinin in polymersome nanoparticles. Physicochemical measurements such as dynamic light scattering, transmission electron microscopy, scanning electron microscopy, and atomic force microscopy confirmed the proper encapsulation of silibinin in nanoparticles. Furthermore, antiproliferative and apoptotic activities of silibinin encapsulated in polymersome nanoparticles (SPNs) on MDA-MB-231 breast cancer cell line were validated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, Annexin V/Propidium Iodide measurement, and cell cycle analysis. In addition, quantitative reverse transcription polymerase chain reaction analysis confirmed that SPNs can repress oncogenic microRNAs (miRNAs) such as miR-125b and miR-182, as well as antiapoptotic genes such as Bcl2. SPNs can also induce overexpression of proapoptotic target genes such as P53, CASP9, and BAX directly and/or indirectly (through regulation of miRNAs). Our results suggested that polymersomes can be used as stable carriers in nano-dimensions and SPNs can be considered as a promising pharmacological agent for cancer therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Apoptosis , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Nanopartículas/química , Polímeros/química , Silibina/uso terapéutico , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , MicroARNs/metabolismo , Nanopartículas/ultraestructura , Tamaño de la Partícula , Silibina/farmacología , Regulación hacia Arriba/efectos de los fármacos
18.
Mol Biol Rep ; 46(5): 5103-5112, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31290055

RESUMEN

The therapeutic application of recombinant proteins is limited due to their inherent structural complexity. Additionally, screening of therapeutic potential of protein products requires an appropriate testing platform to achieve biological relevance. Fabrication of three dimensional cultures bridges the gap between in vitro based monolayer cultures and clinical applications. In this perspective, glioblastoma U-87 MG and breast cancer MCF7 spheroids were generated to assess the therapeutic prospect of recombinant PTEN protein. PTEN bound to silver nanoclusters was encapsulated within PEG coating, which resulted in fabrication of spherical nanocarriers named as PTEN-nanocomposites. Internalization of PTEN-nanocomposites in the spheroids was confirmed by confocal microscopy. Upon uptake, PTEN-nanocomposites led to modulation of cyclins and apoptosis gene regulators culminating in cell cycle arrest and reduced cell viability as confirmed by calcein-AM/PI dual staining and alamar blue assay. Further, combination of tamoxifen and PTEN-nanocomposites on U-87 MG spheroids resulted in two-fold reduction of drug dosage. The study revealed that the monolayer culture results translated to the 3D culture as well, however higher dose of the recombinant PTEN was required for the spheroid system. The anti-proliferative role of PTEN-nanocomposites in a complex 3D environment augments its biological implication and paves the way for recombinant PTEN based therapeutic applications.


Asunto(s)
Fosfohidrolasa PTEN/farmacología , Polietilenglicoles/química , Esferoides Celulares/citología , Tamoxifeno/farmacología , Técnicas de Cultivo de Célula , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Nanopartículas del Metal , Microscopía Confocal , Nanocompuestos , Fosfohidrolasa PTEN/química , Fosfohidrolasa PTEN/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo
19.
Proc Natl Acad Sci U S A ; 113(47): 13318-13323, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27821760

RESUMEN

We report the in vitro long-term (20 wk) changes in cells exposed to well-characterized gold nanoparticles (Au NPs) with varying shapes and surface coatings under both chronic (exposure to Au NPs continuously over 20 wk) and nonchronic (initial acute cell exposure to Au NPs, followed by 20 wk in NP-free cell media) conditions. Both chronic and nonchronic Au NPs exposures at low dose induce modifications at the gene level after long periods. In attempt to overcome from the injuries caused by nanoparticle exposure, genes related to oxidative stress, cell cycle regulation, and inflammation are among those presenting differential expression levels. Surprisingly, the nonchronic exposure induced more gene expression changes than its chronic counterpart and the stress effects caused by this type of exposure were sustained even after 20 wk without any additional NP exposure. NP surface chemistry played an important role in the alteration of gene regulation. Overall, our data suggest that (i) cells can adaptively respond to chronic, low-level NP insults; (ii) the cell stress response is not reversible over time upon removal of NPs upon acute, nonchronic exposure; and (iii) polyethylene glycol is not as benign a surface chemistry as is generally supposed.


Asunto(s)
Fibroblastos/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Oro/toxicidad , Nanopartículas del Metal/química , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Fibroblastos/citología , Regulación de la Expresión Génica/efectos de los fármacos , Oro/química , Oro/farmacología , Humanos , Estrés Oxidativo/efectos de los fármacos , Polietilenglicoles/toxicidad , Pruebas de Toxicidad Aguda , Pruebas de Toxicidad Crónica
20.
Drug Dev Ind Pharm ; 45(3): 423-429, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30449192

RESUMEN

OBJECTIVE: S-nitrosogluthatione (GSNO), a S-nitrosothiol, is a commonly used as nitric oxide (NO•) donor. However, its half-life is too short for a direct therapeutic use. To protect and ensure a sustained release of NO•, the encapsulation of GSNO into nanoparticles may be an interesting option. METHODS: In this work, we have investigated the early (4 h) and late (24 h) transcriptomic response of THP-1 human monocytes cells to two doses (1.4 and 6 µM) of either free or Eudragit® nano-encapsulated GSNO using RNA microarray. RESULTS: After exposure to free GSNO, genes mainly involved in apoptosis, cell differentiation, immune response and metabolic processes were differentially expressed. Although, cells exposed to free or encapsulated GSNO behave differently, activation of genes involved in blood coagulation, immune response and cell cycle was observed in both conditions. CONCLUSIONS: These results suggest that the encapsulation of low doses of GSNO into Eudragit® nanoparticles leads to a progressive release of GSNO making this compound a possible oral therapy for several biomedical applications like inflammatory bowel diseases.


Asunto(s)
S-Nitrosoglutatión/farmacocinética , Transcriptoma/efectos de los fármacos , Apoptosis/efectos de los fármacos , Coagulación Sanguínea/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Semivida , Humanos , Inmunidad/efectos de los fármacos , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Nanopartículas/metabolismo , Óxido Nítrico/metabolismo , Ácidos Polimetacrílicos/química , Células THP-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA