Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.683
Filtrar
1.
Drug Chem Toxicol ; 47(4): 404-415, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38949608

RESUMEN

Although the presence of nitro groups in chemicals can be recognized as structural alerts for mutagenicity and carcinogenicity, nitroaromatic compounds have attracted considerable interest as a class of agents that can serve as source of potential new anticancer agents. In the present study, the in vitro cytotoxicity, genotoxicity, and mutagenicity of three synthetic ortho-nitrobenzyl derivatives (named ON-1, ON-2 and ON-3) were evaluated by employing human breast and ovarian cancer cell lines. A series of biological assays was carried out with and without metabolic activation. Complementarily, computational predictions of the pharmacokinetic properties and druglikeness of the compounds were performed in the Swiss ADME platform. The MTT assay showed that the compounds selectively affected selectively the cell viability of cancer cells in comparison with a nontumoral cell line. Additionally, the metabolic activation enhanced cytotoxicity, and the compounds affected cell survival, as demonstrated by the clonogenic assay. The comet assay, the cytokinesis-block micronucleus assay, and the immunofluorescence of the γ-H2AX foci formation assay have that the compounds caused chromosomal damage to the cancer cells, with and without metabolic activation. The results obtained in the present study showed that the compounds assessed were genotoxic and mutagenic, inducing double-strand breaks in the DNA structure. The high selectivity indices observed for the compounds ON-2 and ON-3, especially after metabolic activation with the S9 fraction, must be highlighted. These experimental biological results, as well as the theoretical properties predicted for the compounds have shown that they are promising anticancer candidates to be exploited in additional studies.


Asunto(s)
Activación Metabólica , Antineoplásicos , Supervivencia Celular , Daño del ADN , Humanos , Supervivencia Celular/efectos de los fármacos , Antineoplásicos/toxicidad , Antineoplásicos/farmacología , Antineoplásicos/química , Daño del ADN/efectos de los fármacos , Línea Celular Tumoral , Pruebas de Micronúcleos , Mutágenos/toxicidad , Ensayo Cometa , Pruebas de Mutagenicidad , Femenino , Nitrobencenos/toxicidad , Nitrobencenos/química , Neoplasias de la Mama/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias Ováricas/patología , Neoplasias Ováricas/tratamiento farmacológico , Relación Dosis-Respuesta a Droga
2.
J Biochem Mol Toxicol ; 38(7): e23764, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38963172

RESUMEN

Obesity is an established risk factor for numerous malignancies, although it remains uncertain whether the disease itself or weight-loss drugs are responsible for a greater predisposition to cancer. The objective of the current study was to determine the impact of dulaglutide on genetic and epigenetic DNA damage caused by obesity, which is a crucial factor in the development of cancer. Mice were administered a low-fat or high-fat diet for 12 weeks, followed by a 5-week treatment with dulaglutide. Following that, modifications of the DNA bases were examined using the comet assay. To clarify the underlying molecular mechanisms, oxidized and methylated DNA bases, changes in the redox status, levels of inflammatory cytokines, and the expression levels of some DNA repair genes were evaluated. Animals fed a high-fat diet exhibited increased body weights, elevated DNA damage, oxidation of DNA bases, and DNA hypermethylation. In addition, obese mice showed altered inflammatory responses, redox imbalances, and repair gene expressions. The findings demonstrated that dulaglutide does not exhibit genotoxicity in the investigated conditions. Following dulaglutide administration, animals fed a high-fat diet demonstrated low DNA damage, less oxidation and methylation of DNA bases, restored redox balance, and improved inflammatory responses. In addition, dulaglutide treatment restored the upregulated DNMT1, Ogg1, and p53 gene expression. Overall, dulaglutide effectively maintains DNA integrity in obese animals. It reduces oxidative DNA damage and hypermethylation by restoring redox balance, modulating inflammatory responses, and recovering altered gene expressions. These findings demonstrate dulaglutide's expediency in treating obesity and its associated complications.


Asunto(s)
Daño del ADN , Metilación de ADN , Reparación del ADN , Dieta Alta en Grasa , Péptidos Similares al Glucagón , Fragmentos Fc de Inmunoglobulinas , Oxidación-Reducción , Proteínas Recombinantes de Fusión , Animales , Péptidos Similares al Glucagón/análogos & derivados , Péptidos Similares al Glucagón/farmacología , Metilación de ADN/efectos de los fármacos , Fragmentos Fc de Inmunoglobulinas/farmacología , Daño del ADN/efectos de los fármacos , Ratones , Reparación del ADN/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Proteínas Recombinantes de Fusión/farmacología , Masculino , Oxidación-Reducción/efectos de los fármacos , Inflamación/metabolismo , Inflamación/genética , Estrés Oxidativo/efectos de los fármacos , Obesidad/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/genética , Regulación de la Expresión Génica/efectos de los fármacos , Ratones Endogámicos C57BL
3.
Arh Hig Rada Toksikol ; 75(2): 137-146, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38963137

RESUMEN

Traditional medicine has used sage (Salvia officinalis L.) preparations for centuries to prevent and treat various inflammatory and oxidative stress-induced conditions. The aim of this in vitro study was to determine the bioactive properties of a sage leave extract obtained with environmentally friendly aqueous extraction and lyophilisation in primary human peripheral blood cells. To that end we measured the total phenolic and flavonoid content (TPC and TFC, respectively) with gas chromatography-mass spectrometry (GC-MS). Non-cytotoxic concentrations determined with the trypan blue assay were used to assess the antioxidant (DPPH, ABTS, and PAB assay), antigenotoxic (CBMN assay), immunomodulatory (IL-1ß and TNF-α), and neuroprotective effects (AChE inhibition). The extract contained high TPC (162 mg GAE/g of dry extract) and TFC (39.47 mg QE/g of dry extract) concentrations, while ß-thujone content was unexpectedly low (below 0.9 %). Strong radical-scavenging activity combined with glutathione reductase activation led to a decrease in basal and H2O2-induced oxidative stress and DNA damage. A decrease in TNF-α and increase in IL-1ß levels suggest complex immunomodulatory response that could contribute to antioxidant and, together with mild AChE inhibition, neuroprotective effects. Overall, this study has demonstrated that aqueous sage leave extract reduces the levels of thujone, 1,8-cineole, pinene, and terpene ketones that could be toxic in high concentrations, while maintaining high concentrations of biologically active protective compounds which have a potential to prevent and/or treat inflammatory and oxidative stress-related conditions.


Asunto(s)
Inflamación , Leucocitos Mononucleares , Estrés Oxidativo , Extractos Vegetales , Salvia officinalis , Humanos , Extractos Vegetales/farmacología , Extractos Vegetales/química , Leucocitos Mononucleares/efectos de los fármacos , Salvia officinalis/química , Inflamación/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Antioxidantes/farmacología , Daño del ADN/efectos de los fármacos , Hojas de la Planta/química
4.
Sci Rep ; 14(1): 15506, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38969725

RESUMEN

Relatively low levels of antioxidant enzymes coupled with high oxygen metabolism result in the formation of numerous oxidative DNA damages in the tissues of the central nervous system. Recently, kynurenic acid (KYNA), knowns for its neuroprotective properties, has gained increasing attention in this context. Therefore, our hypothesis assumed that increased KYNA levels in the brain would positively influence mRNA expression of selected enzymes of the base excision repair pathway as well as enhance their efficiency in excising damaged nucleobases in specific areas of the sheep brain. The study was conducted on adult anestrous sheep (n = 18), in which two different doses of KYNA (20 and 100 µg/day) were infused into the third brain ventricle for three days. Molecular and biochemical analysis included the hypothalamus (preoptic and mediol-basal areas), hippocampus (CA3 field) and amygdala (central amygdaloid nucleus), dissected from the brain of sheep euthanized immediately after the last infusion. The results revealed a significant increase P < 0.001) in the relative mRNA abundance of N-methylpurine DNA glycosylase (MPG) following administration of both dose of KYNA across all examined tissues. The transcription of thymine-DNA glycosylase (TDG) increased significantly (P < 0.001) in all tissues in response to the lower KYNA dose compared to the control group. Moreover, 8-oxoguanine (8-oxoG) DNA glycosylase (OGG1) mRNA levels were also higher in both animal groups (P < 0.001). In addition, in the hypothalamus, hippocampus and amygdala, AP endonuclease 1 (APE1) mRNA expression increased under both doses of KYNA. Moreover, the both dose of KYNA significantly stimulated the efficiency of 8-oxoG excision in hypothalamus and amygdala (P < 0.05-0.001). The lower and higher doses of KYNA significantly influenced the effectiveness of εA and εC in all structures (P < 0.01-0.001). In conclusion, the favorable effect of KYNA in the brain may include the protection of genetic material in nerve and glial cells by stimulating the expression and efficiency of BER pathway enzymes.


Asunto(s)
Encéfalo , ADN Glicosilasas , Reparación del ADN , Ácido Quinurénico , Animales , Reparación del ADN/efectos de los fármacos , Ovinos , Ácido Quinurénico/metabolismo , ADN Glicosilasas/metabolismo , ADN Glicosilasas/genética , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Hipotálamo/metabolismo , Hipotálamo/efectos de los fármacos , ARN Mensajero/metabolismo , ARN Mensajero/genética , Daño del ADN/efectos de los fármacos , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Femenino , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Reparación por Escisión
5.
Biol Res ; 57(1): 44, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38965573

RESUMEN

BACKGROUND: Exposure of humans and animals to heavy metals is increasing day-by-day; thus, lead even today remains of significant public health concern. According to CDC, blood lead reference value (BLRV) ranges from 3.5 µg/dl to 5 µg/dl in adults. Recently, almost 2.6% decline in male fertility per year has been reported but the cause is not well established. Lead (Pb2+) affects the size of testis, semen quality, and secretory functions of prostate. But the molecular mechanism(s) of lead toxicity in sperm cells is not clear. Thus, present study was undertaken to evaluate the adverse effects of lead acetate at environmentally relevant exposure levels (0.5, 5, 10 and 20 ppm) on functional and molecular dynamics of spermatozoa of bucks following in vitro exposure for 15 min and 3 h. RESULTS: Lead significantly decreased motility, viable count, and motion kinematic patterns of spermatozoa like curvilinear velocity, straight-line velocity, average path velocity, beat cross frequency and maximum amplitude of head lateral displacement even at 5 ppm concentration. Pb2+ modulated intracellular cAMP and Ca2+ levels in sperm cells through L-type calcium channels and induced spontaneous or premature acrosome reaction (AR) by increasing tyrosine phosphorylation of sperm proteins and downregulated mitochondrial transmembrane potential. Lead significantly increased DNA damage and apoptosis as well. Electron microscopy studies revealed Pb2+ -induced deleterious effects on plasma membrane of head and acrosome including collapsed cristae in mitochondria. CONCLUSIONS: Pb2+ not only mimics Ca2+ but also affects cellular targets involved in generation of cAMP, mitochondrial transmembrane potential, and ionic exchange. Lead seems to interact with Ca2+ channels because of charge similarity and probably enters the sperm cell through these channels and results in hyperpolarization. Our findings also indicate lead-induced TP and intracellular Ca2+ release in spermatozoa which in turn may be responsible for premature acrosome exocytosis which is essential feature of capacitation for fertilization. Thus, lead seems to reduce the fertilizing capacity of spermatozoa even at 0.5 ppm concentrations.


Asunto(s)
Reacción Acrosómica , Acrosoma , Calcio , Plomo , Motilidad Espermática , Espermatozoides , Masculino , Espermatozoides/efectos de los fármacos , Calcio/metabolismo , Motilidad Espermática/efectos de los fármacos , Animales , Acrosoma/efectos de los fármacos , Plomo/toxicidad , Reacción Acrosómica/efectos de los fármacos , AMP Cíclico/metabolismo , Bovinos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Análisis de Semen , Daño del ADN/efectos de los fármacos , Compuestos Organometálicos/toxicidad , Compuestos Organometálicos/farmacología
6.
Sci Rep ; 14(1): 15314, 2024 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961104

RESUMEN

This work examines the capacity of Naringin and Rutin to influence the DNA damage response (DDR) pathway by investigating their interactions with key DDR proteins, including PARP-1, ATM, ATR, CHK1, and WEE1. Through a combination of in silico molecular docking and in vitro evaluations, we investigated the cytotoxic and genotoxic effects of these compounds on MDA-MB-231 cells, comparing them to normal human fibroblast cells (2DD) and quiescent fibroblast cells (QFC). The research found that Naringin and Rutin had strong affinities for DDR pathway proteins, indicating their capacity to specifically regulate DDR pathways in cancer cells. Both compounds exhibited preferential cytotoxicity towards cancer cells while preserving the vitality of normal 2DD fibroblast cells, as demonstrated by cytotoxicity experiments conducted at a dose of 10 µM. The comet experiments performed particularly on QFC cells provide valuable information on the genotoxic impact of Naringin and Rutin, highlighting the targeted initiation of DNA damage in cancer cells. The need to use precise cell models to appropriately evaluate toxicity and genotoxicity is emphasized by this discrepancy. In addition, ADMET and drug-likeness investigations have emphasized the pharmacological potential of these compounds; however, they have also pointed out the necessity for optimization to improve their therapeutic profiles. The antioxidant capabilities of Naringin and Rutin were assessed using DPPH and free radical scavenging assays at a concentration of 10 µM. The results confirmed that both compounds have a role in reducing oxidative stress, hence enhancing their anticancer effects. Overall, Naringin and Rutin show potential as medicines for modulating the DDR in cancer treatment. They exhibit selective toxicity towards cancer cells while sparing normal cells and possess strong antioxidant properties. This analysis enhances our understanding of the therapeutic uses of natural chemicals in cancer treatment, supporting the need for more research on their mechanisms of action and clinical effectiveness.


Asunto(s)
Antioxidantes , Neoplasias de la Mama , Daño del ADN , Flavanonas , Simulación del Acoplamiento Molecular , Estrés Oxidativo , Rutina , Humanos , Flavanonas/farmacología , Rutina/farmacología , Daño del ADN/efectos de los fármacos , Antioxidantes/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Estrés Oxidativo/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Supervivencia Celular/efectos de los fármacos
7.
Biol Pharm Bull ; 47(7): 1255-1264, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38972750

RESUMEN

Traditional Chinese Medicine, known for its minimal side effects and significant clinical efficacy, has attracted considerable interest for its potential in cancer therapy. In particular, Inula helenium L. has demonstrated effectiveness in inhibiting a variety of cancers. This study focuses on alantolactone (ALT), a prominent compound from Inula helenium L., recognized for its anti-cancer capabilities across multiple cancer types. The primary objective of this study is to examine the influence of ALT on the proliferation, apoptosis, cell cycle, and tumor growth of cervical cancer (CC) cells, along with its associated signaling pathways. To determine protein expression alterations, Western blot analysis was conducted. Furthermore, an in vivo model was created by subcutaneously injecting HeLa cells into nude mice to assess the impact of ALT on cervical cancer. Our research thoroughly investigates the anti-tumor potential of ALT in the context of CC. ALT was found to inhibit cell proliferation and induce apoptosis in SiHa and HeLa cell lines, particularly targeting ataxia-telangiectasia mutated (ATM) proteins associated with DNA damage. The suppression of DNA damage and apoptosis induction when ATM was inhibited underscores the crucial role of the ATM/cell cycle checkpoint kinase 2 (CHK2) axis in ALT's anti-tumor effects. In vivo studies with a xenograft mouse model further validated ALT's effectiveness in reducing CC tumor growth and promoting apoptosis. This study offers new insights into how ALT combats CC, highlighting its promise as an effective anti-cervical cancer agent and providing hope for improved treatment outcomes for CC patients.


Asunto(s)
Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada , Quinasa de Punto de Control 2 , Daño del ADN , Lactonas , Ratones Desnudos , Sesquiterpenos de Eudesmano , Transducción de Señal , Neoplasias del Cuello Uterino , Humanos , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/metabolismo , Apoptosis/efectos de los fármacos , Femenino , Quinasa de Punto de Control 2/metabolismo , Daño del ADN/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sesquiterpenos de Eudesmano/farmacología , Sesquiterpenos de Eudesmano/uso terapéutico , Lactonas/farmacología , Lactonas/uso terapéutico , Células HeLa , Proliferación Celular/efectos de los fármacos , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/uso terapéutico , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos BALB C , Ratones , Inula/química
8.
Sci Rep ; 14(1): 14484, 2024 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-38914713

RESUMEN

Among the myriad of nanoparticles, silica nanoparticles (SiO2NPs) have gained significant attention since they are extensively produced and used across several kinds of industries. Because of its widespread usage, there has been increasing concern about the potential health effects. This study aims to evaluate the effects of SiO2NPs on Interleukin-6 (IL-6) gene expression in human lung epithelial cell lines (A549). In this study, A549 cells were exposed to SiO2NPs at concentrations of 0, 1, 10, 50, 100, and 200 µg/mL for 24 and 48 h. The IL-6 gene expression was assessed using Real-Time RT-PCR. Additionally, the impact of SiO2NPs on the viability of A549 cells was determined by MTT assay. Statistical analysis was performed using GraphPad Prism software 8.0. MTT assay results indicated a concentration-dependent impact on cell survival. After 24 h, survival decreased from 80 to 68% (1-100 µg/mL), rising to 77% at higher concentrations. After 48 h, survival dropped from 97 to 80%, decreasing to 90% at higher concentrations. RT-PCR showed a dose-response relationship in cellular toxicity up to 10 µg/mL. At higher concentrations, there was increased IL-6 gene expression, mitigating SiO2NP-induced cytotoxic effects. The study shows that the viability and proliferation of A549 cells are impacted by different SiO2NPs concentrations. There may be a potential correlation between IL-6 gene expression reduction and a mechanism linked to cellular toxicity. However, at higher concentrations, an unknown mechanism increases IL-6 gene expression, reducing SiO2NPs' cytotoxic effects. These effects are concentration-dependent and not influenced by exposure times. Further investigation is recommended to determine this mechanism's nature and implications, particularly in cancer research.


Asunto(s)
Supervivencia Celular , Interleucina-6 , Nanopartículas , Dióxido de Silicio , Humanos , Dióxido de Silicio/toxicidad , Dióxido de Silicio/química , Células A549 , Nanopartículas/toxicidad , Nanopartículas/química , Interleucina-6/metabolismo , Interleucina-6/genética , Supervivencia Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos
9.
J Hazard Mater ; 475: 134900, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38878440

RESUMEN

The expected increments in the production/use of bioplastics, as an alternative to petroleum-based plastics, require a deep understanding of their potential environmental and health hazards, mainly as nanoplastics (NPLs). Since one important exposure route to NPLs is through inhalation, this study aims to determine the fate and effects of true-to-life polylactic acid nanoplastics (PLA-NPLs), using the in vitro Calu-3 model of bronchial epithelium, under air-liquid interphase exposure conditions. To determine the harmful effects of PLA-NPLs in a more realistic scenario, both acute (24 h) and long-term (1 and 2 weeks) exposures were used. Flow cytometry results indicated that PLA-NPLs internalized easily in the barrier (∼10 % at 24 h and ∼40 % after 2 weeks), which affected the expression of tight-junctions formation (∼50 % less vs control) and the mucus secretion (∼50 % more vs control), both measured by immunostaining. Interestingly, significant genotoxic effects (DNA breaks) were detected by using the comet assay, with long-term effects being more marked than acute ones (7.01 vs 4.54 % of DNA damage). When an array of cellular proteins including cytokines, chemokines, and growth factors were used, a significant over-expression was mainly found in long-term exposures (∼20 proteins vs 5 proteins after acute exposure). Overall, these results described the potential hazards posed by PLA-NPLs, under relevant long-term exposure scenarios, highlighting the advantages of the model used to study bronchial epithelium tissue damage, and signaling endpoints related to inflammation.


Asunto(s)
Poliésteres , Poliésteres/toxicidad , Poliésteres/química , Humanos , Línea Celular , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Citocinas/metabolismo , Microplásticos/toxicidad , Daño del ADN/efectos de los fármacos , Nanopartículas/toxicidad , Nanopartículas/química , Epitelio/efectos de los fármacos , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Células Epiteliales/efectos de los fármacos , Uniones Estrechas/efectos de los fármacos
10.
J Control Release ; 371: 371-385, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38849089

RESUMEN

The efficacy of DNA-damaging agents, such as the topoisomerase I inhibitor SN38, is often compromised by the robust DNA repair mechanisms in tumor cells, notably homologous recombination (HR) repair. Addressing this challenge, we introduce a novel nano-strategy utilizing binary tumor-killing mechanisms to enhance the therapeutic impact of DNA damage and mitochondrial dysfunction in cancer treatment. Our approach employs a synergistic drug pair comprising SN38 and the BET inhibitor JQ-1. We synthesized two prodrugs by conjugating linoleic acid (LA) to SN38 and JQ-1 via a cinnamaldehyde thioacetal (CT) bond, facilitating co-delivery. These prodrugs co-assemble into a nanostructure, referred to as SJNP, in an optimal synergistic ratio. SJNP was validated for its efficacy at both the cellular and tissue levels, where it primarily disrupts the transcription factor protein BRD4. This disruption leads to downregulation of BRCA1 and RAD51, impairing the HR process and exacerbating DNA damage. Additionally, SJNP releases cinnamaldehyde (CA) upon CT linkage cleavage, elevating intracellular ROS levels in a self-amplifying manner and inducing ROS-mediated mitochondrial dysfunction. Our results indicate that SJNP effectively targets murine triple-negative breast cancer (TNBC) with minimal adverse toxicity, showcasing its potential as a formidable opponent in the fight against cancer.


Asunto(s)
Acroleína , Camptotecina , Sistemas de Liberación de Medicamentos , Nanopartículas , Neoplasias de la Mama Triple Negativas , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Nanopartículas/administración & dosificación , Nanopartículas/química , Animales , Humanos , Femenino , Línea Celular Tumoral , Acroleína/análogos & derivados , Acroleína/administración & dosificación , Acroleína/química , Camptotecina/análogos & derivados , Camptotecina/administración & dosificación , Camptotecina/uso terapéutico , Camptotecina/farmacología , Profármacos/administración & dosificación , Profármacos/uso terapéutico , Ácido Linoleico/química , Ácido Linoleico/administración & dosificación , Triazoles/administración & dosificación , Triazoles/farmacología , Triazoles/química , Daño del ADN/efectos de los fármacos , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Ratones Desnudos , Ratones , Proteínas de Ciclo Celular/metabolismo , Factores de Transcripción/metabolismo , Inhibidores de Topoisomerasa I/administración & dosificación , Proteínas que Contienen Bromodominio , Azepinas
11.
Drug Dev Ind Pharm ; 50(6): 561-575, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38832870

RESUMEN

INTRODUCTION: Breast cancer (BC) is the most common malignancy in women globally. Significant progress has been made in developing structural nanoparticles (NPs) and formulations for targeted smart drug delivery (SDD) of pharmaceuticals, improving the precision of tumor cell targeting in therapy. SIGNIFICANCE: Magnetic hyperthermia (MHT) treatment using magneto-liposomes (MLs) has emerged as a promising adjuvant cancer therapy. METHODS: CoFe2O4 magnetic NPs (MNPs) were conjugated with nanoliposomes to form MLs, and the anticancer drug quercetin (Que) was loaded into MLs, forming Que-MLs composites for antitumor approach. The aim was to prepare Que-MLs for DD systems (DDS) under an alternating magnetic field (AMF), termed chemotherapy/hyperthermia (chemo-HT) techniques. The encapsulation efficiency (EE), drug loading capacity (DL), and drug release (DR) of Que and Que-MLs were evaluated. RESULTS: The results confirmed successful Que-loading on the surface of MLs, with an average diameter of 38 nm and efficient encapsulation into MLs (69%). In vitro, experimental results on MCF-7 breast cells using MHT showed high cytotoxic effects of novel Que-MLs on MCF-7 cells. Various analyses, including cytotoxicity, apoptosis, cell migration, western blotting, fluorescence imaging, and cell membrane internalization, were conducted. The Acridine Orange-ethidium bromide double fluorescence test identified 35% early and 55% late apoptosis resulting from Que-MLs under the chemo-HT group. TEM results indicated MCF-7 cell membrane internalization and digestion of Que-MLs, suggesting the presence of early endosome-like vesicles on the cytoplasmic periphery. CONCLUSIONS: Que-MLs exhibited multi-modal chemo-HT effects, displaying high toxicity against MCF-7 BC cells and showing promise as a potent cytotoxic agent for BC chemotherapy.


Asunto(s)
Apoptosis , Neoplasias de la Mama , Daño del ADN , Hipertermia Inducida , Liposomas , Quercetina , Humanos , Quercetina/farmacología , Quercetina/administración & dosificación , Quercetina/química , Células MCF-7 , Apoptosis/efectos de los fármacos , Hipertermia Inducida/métodos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Daño del ADN/efectos de los fármacos , Cobalto/química , Cobalto/administración & dosificación , Cobalto/farmacología , Femenino , Compuestos Férricos/química , Liberación de Fármacos , Antineoplásicos/farmacología , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas de Magnetita/química , Supervivencia Celular/efectos de los fármacos , Campos Magnéticos
12.
Redox Biol ; 74: 103224, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38865904

RESUMEN

BACKGROUND: Silicosis, characterized by interstitial lung inflammation and fibrosis, poses a significant health threat. ATII cells play a crucial role in alveolar epithelial repair and structural integrity maintenance. Inhibiting ATII cell senescence has shown promise in silicosis treatment. However, the mechanism behind silica-induced senescence remains elusive. METHODS: The study employed male C57BL/6 N mice and A549 human alveolar epithelial cells to investigate silicosis and its potential treatment. Silicosis was induced in mice via intratracheal instillation of crystalline silica particles, with honokiol administered intraperitoneally for 14 days. Silica-induced senescence in A549 cells was confirmed, and SIRT3 knockout and overexpression cell lines were generated. Various analyses were conducted, including immunoblotting, qRT-PCR, histology, and transmission electron microscopy. Statistical significance was determined using one-way ANOVA with Tukey's post-hoc test. RESULTS: This study elucidates how silica induces ATII cell senescence, emphasizing mtDNA damage. Notably, honokiol (HKL) emerges as a promising anti-senescence and anti-fibrosis agent, acting through sirt3. honokiol effectively attenuated senescence in ATII cells, dependent on sirt3 expression, while mitigating mtDNA damage. Sirt3, a class III histone deacetylase, regulates senescence and mitochondrial stress. HKL activates sirt3, protecting against pulmonary fibrosis and mitochondrial damage. Additionally, HKL downregulated cGAS expression in senescent ATII cells induced by silica, suggesting sirt3's role as an upstream regulator of the cGAS/STING signaling pathway. Moreover, honokiol treatment inhibited the activation of the NF-κB signaling pathway, associated with reduced oxidative stress and mtDNA damage. Notably, HKL enhanced the activity of SOD2, crucial for mitochondrial function, through sirt3-mediated deacetylation. Additionally, HKL promoted the deacetylation activity of sirt3, further safeguarding mtDNA integrity. CONCLUSIONS: This study uncovers a natural compound, HKL, with significant anti-fibrotic properties through activating sirt3, shedding light on silicosis pathogenesis and treatment avenues.


Asunto(s)
Células Epiteliales Alveolares , Compuestos de Bifenilo , Senescencia Celular , Lignanos , Transducción de Señal , Silicosis , Sirtuina 3 , Animales , Silicosis/metabolismo , Silicosis/tratamiento farmacológico , Silicosis/patología , Silicosis/etiología , Sirtuina 3/metabolismo , Sirtuina 3/genética , Senescencia Celular/efectos de los fármacos , Ratones , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Compuestos de Bifenilo/farmacología , Humanos , Lignanos/farmacología , Transducción de Señal/efectos de los fármacos , Masculino , Células A549 , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Modelos Animales de Enfermedad , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Daño del ADN/efectos de los fármacos , Compuestos Alílicos , Fenoles
13.
Nanoscale ; 16(26): 12502-12509, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38873939

RESUMEN

Targeted protein degradation through PROteolysis TArgeting Chimeras (PROTACs) is a relatively new modality in cellular interventions. The minimum requirement for PROTACs to function is forming a tertiary complex of the protein of interest (POI), E3 ligase, and the molecular glue PROTAC. Here, we propose a new approach to modulate the nano-environment interactome of a non-protein target through a plausible quaternary complex of interactome-biomolecule of interest (BOI)-PROTAC and E3 ligase. We report nucleic acid-targeting PROTAC (NA-TAC) molecules by conjugating DNA-binding and E3 ligase ligands. We demonstrate that NA-TACs can target the G-quadruplex DNA and induce elevated DNA damage and cytotoxicity compared to the conventional G-quadruplex binding ligands. Our new class of NA-TACs lays the foundation for small molecule-based non-protein targeting PROTACs for interactome and nanoenvironment mapping and nucleic acid-targeted precision medicines.


Asunto(s)
Antineoplásicos , G-Cuádruplex , Proteolisis , Ubiquitina-Proteína Ligasas , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Proteolisis/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , G-Cuádruplex/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Ligandos , Ácidos Nucleicos/química , Ácidos Nucleicos/metabolismo , ADN/química , ADN/metabolismo , Quimera Dirigida a la Proteólisis
14.
In Vivo ; 38(4): 1719-1730, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38936939

RESUMEN

BACKGROUND/AIM: ClFdA is a second-generation antineoplastic agent that has demonstrated significant anticancer activity, particularly against acute lymphoblastic leukemia and has been shown to have radiosensitizing activity. The aim of the study was to explore the genotoxic, cytotoxic and radiosensitizing effects of clofarabine (ClFdA) on bone marrow cells (BMCs), normoblasts and leukocytes of mice in vivo. MATERIALS AND METHODS: Cytotoxicity was determined by the reduction in reticulocytes (RET), and genotoxicity was determined by the induction of micronucleated reticulocytes (MN-RET) in the peripheral blood and by DNA break induction in leukocytes determined by single-cell gel electrophoresis (SCGE). The radiosensitizing capacity of ClFdA was determined in leukocytes and BMCs by SCGE. RESULTS: Two mechanisms of MN-RET induction were identified according to the antecedents, that could be due to inhibition of DNA synthesis and demethylation of G-C regions, and subsequent chromosome fragility. ClFdA cytotoxicity causes two contiguous peaks, an early peak that seems to inhibit MN-RET induction and a second peak that seems to be caused by ribonucleotide reductase (RR) and/or DNA synthesis inhibitions. ClFdA induced early DNA damage in noncycling leukocytes, and also radiosensitizes leukocytes immediately after treatment. ClFdA-ionizing radiation (IR) causes two time-dependent episodes of DNA damage, the latest after 80 min triggers a major breakage of DNA. In terms of the number of damaged cells, leukocytes and BMCs are similarly sensitive to ionizing radiation; BMCs are slightly more sensitive than leukocytes to ClFdA, but BMCs are doubly sensitive to combined treatment. CONCLUSION: ClFdA causes early DNA damage and radiosensitivity in non-proliferating leukocytes, which rules out the most favored hypotheses of the participation of RR and DNA polymerase inhibition.


Asunto(s)
Clofarabina , Daño del ADN , Leucocitos , Fármacos Sensibilizantes a Radiaciones , Animales , Clofarabina/farmacología , Ratones , Fármacos Sensibilizantes a Radiaciones/farmacología , Leucocitos/efectos de los fármacos , Leucocitos/efectos de la radiación , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Arabinonucleósidos/farmacología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/efectos de la radiación , Células de la Médula Ósea/metabolismo , Nucleótidos de Adenina/farmacología , Masculino , Reticulocitos/efectos de los fármacos , Reticulocitos/efectos de la radiación , Antineoplásicos/farmacología , Pruebas de Micronúcleos
15.
Aging (Albany NY) ; 16(11): 9547-9557, 2024 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-38829772

RESUMEN

Neratinib, a typical small-molecule, pan-human tyrosine kinase inhibitor (TKI), has been licensed for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer. However, the underlying pharmacological mechanism is still unknown. In the current study, we report a novel function of Neratinib by showing that its treatment stimulates senescence of the mammary cancer AU565 cells. Our results demonstrate that Neratinib induces mitochondrial injury by increasing mitochondrial reactive oxygen species (ROS) and reducing intracellular adenosine triphosphate (ATP). Also, we found that Neratinib induced DNA damage by increasing the levels of 8-Hydroxy-desoxyguanosine (8-OHdG) and γH2AX in AU565 cells. Additionally, Neratinib reduced the levels of telomerase activity after 7 and 14 days incubation. Importantly, the senescence-associated-ß-galactosidase (SA-ß-Gal) assay revealed that Neratinib stimulated senescence of AU565 cells. Neratinib decreased the gene levels of human telomerase reverse transcriptase (hTERT) but increased those of telomeric repeat-binding factor 2 (TERF2) in AU565 cells. Further study displayed that Neratinib upregulated the expression of K382 acetylation of p53 (ac-K382) and p21 but reduced the levels of sirtuin-1 (SIRT1). However, overexpression of SIRT1 abolished the effects of Neratinib in cellular senescence. These findings provide strong preclinical evidence of Neratinib's treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama , Senescencia Celular , Quinolinas , Sirtuina 1 , Humanos , Sirtuina 1/metabolismo , Sirtuina 1/genética , Senescencia Celular/efectos de los fármacos , Quinolinas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Daño del ADN/efectos de los fármacos , Telomerasa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Antineoplásicos/farmacología
16.
Biochem Biophys Res Commun ; 725: 150266, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-38878759

RESUMEN

Cisplatin (CDDP) is a platinum-based anticancer drug widely prescribed for its effectiveness in treating various forms of cancer. However, its major side effect is nephrotoxicity. Although several methods have been developed to mitigate CDDP-induced nephrotoxicity, an optimal approach has yet to be established. This study aimed to investigate the "chronotoxicity" of CDDP as a potential strategy to reduce its side effects. Male ICR mice were treated with CDDP (20 mg/kg, intraperitoneal injection, one shot) at zeitgeber time (ZT) 2 or ZT14 (light or dark phase). After 72 h, we collected plasma and kidney and evaluated several markers. We found that body weight change between ZT2 and ZT14 by CDDP was comparable. In contrast, many toxicological factors, such as plasma blood urine nitrogen, plasma creatinine, renal oxidative stress (malondialdehyde), DNA damage (γH2AX), acute kidney injury biomarker (KIM-1), and inflammation (Tnfα), were significantly induced at ZT14 compared to than that of ZT2. Our present data suggested that chronotoxicology might provide beneficial information on the importance of administration timings for toxic evaluations and unacceptable side effects.


Asunto(s)
Antineoplásicos , Ritmo Circadiano , Cisplatino , Riñón , Ratones Endogámicos ICR , Animales , Cisplatino/toxicidad , Masculino , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Antineoplásicos/toxicidad , Antineoplásicos/efectos adversos , Ratones , Ritmo Circadiano/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/metabolismo , Enfermedades Renales/patología
17.
Nutrients ; 16(12)2024 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-38931171

RESUMEN

Taurine, a non-proteogenic amino acid and commonly used nutritional supplement, can protect various tissues from degeneration associated with the action of the DNA-damaging chemotherapeutic agent cisplatin. Whether and how taurine protects human ovarian cancer (OC) cells from DNA damage caused by cisplatin is not well understood. We found that OC ascites-derived cells contained significantly more intracellular taurine than cell culture-modeled OC. In culture, elevation of intracellular taurine concentration to OC ascites-cell-associated levels suppressed proliferation of various OC cell lines and patient-derived organoids, reduced glycolysis, and induced cell protection from cisplatin. Taurine cell protection was associated with decreased DNA damage in response to cisplatin. A combination of RNA sequencing, reverse-phase protein arrays, live-cell microscopy, flow cytometry, and biochemical validation experiments provided evidence for taurine-mediated induction of mutant or wild-type p53 binding to DNA, activation of p53 effectors involved in negative regulation of the cell cycle (p21), and glycolysis (TIGAR). Paradoxically, taurine's suppression of cell proliferation was associated with activation of pro-mitogenic signal transduction including ERK, mTOR, and increased mRNA expression of major DNA damage-sensing molecules such as DNAPK, ATM and ATR. While inhibition of ERK or p53 did not interfere with taurine's ability to protect cells from cisplatin, suppression of mTOR with Torin2, a clinically relevant inhibitor that also targets DNAPK and ATM/ATR, broke taurine's cell protection. Our studies implicate that elevation of intracellular taurine could suppress cell growth and metabolism, and activate cell protective mechanisms involving mTOR and DNA damage-sensing signal transducti.


Asunto(s)
Cisplatino , Daño del ADN , Neoplasias Ováricas , Serina-Treonina Quinasas TOR , Taurina , Proteína p53 Supresora de Tumor , Taurina/farmacología , Humanos , Serina-Treonina Quinasas TOR/metabolismo , Femenino , Neoplasias Ováricas/metabolismo , Daño del ADN/efectos de los fármacos , Cisplatino/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Glucólisis/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Antineoplásicos/farmacología
18.
Cell Physiol Biochem ; 58(3): 273-287, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38881348

RESUMEN

BACKGROUND/AIMS: Inhaled particulate air pollution is associated with cardiotoxicity with underlying mechanisms including oxidative stress and inflammation. Carnosol, commonly found in rosemary and sage, is known to possess a broad range of therapeutic properties such as antioxidant, anti-inflammatory and antiapoptotic. However, its cardioprotective effects on diesel exhaust particles (DEPs)-induced toxicity have not been studied yet. Hence, we evaluated the potential ameliorative effects of carnosol on DEPs-induced heart toxicity in mice, and the underlying mechanisms involved. METHODS: Mice were intratracheally instilled with DEPs (1 mg/kg) or saline, and 1 hour prior to instillation they were given intraperitoneally either carnosol (20 mg/kg) or saline. Twenty-four hours after the DEPs instillation, multiple parameters were evaluated in the heart by enzyme-linked immunosorbent assay, colorimetric assay, Comet assay and Western blot technique. RESULTS: Carnosol has significantly reduced the elevation in the plasma levels of lactate hydrogenase and brain natriuretic peptide induced by DEPs. Likewise, the augmented cardiac levels of proinflammatory cytokines, lipid peroxidation, and total nitric oxide in DEPs-treated groups were significantly normalized with the treatment of carnosol. Moreover, carnosol has markedly reduced the heart mitochondrial dysfunction, as well as DNA damage and apoptosis of mice treated with DEPs. Similarly, carnosol significantly reduced the elevated expressions of phosphorylated nuclear factor-кB (NF-кB) and mitogen-activated protein kinases (MAPKs) in the hearts. Furthermore, the treatment with carnosol has restored the decrease in the expression of sirtuin-1 in the hearts of mice exposed to DEPs. CONCLUSION: Carnosol significantly attenuated DEP-induced cardiotoxicity in mice by suppressing inflammation, oxidative stress, DNA damage, and apoptosis, at least partly via mechanisms involving sirtuin-1 activation and the inhibition of NF-кB and MAPKs activation.


Asunto(s)
Abietanos , Cardiotoxicidad , FN-kappa B , Estrés Oxidativo , Emisiones de Vehículos , Animales , Ratones , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Emisiones de Vehículos/toxicidad , Abietanos/farmacología , Abietanos/uso terapéutico , Masculino , Cardiotoxicidad/etiología , Cardiotoxicidad/metabolismo , Cardiotoxicidad/prevención & control , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxicidad/patología , Estrés Nitrosativo/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Inflamación/inducido químicamente , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Cardiotónicos/farmacología , Cardiotónicos/uso terapéutico , Sirtuina 1/metabolismo , Sirtuina 1/genética , Daño del ADN/efectos de los fármacos
19.
PLoS Negl Trop Dis ; 18(6): e0012251, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38885188

RESUMEN

Fasciolosis caused by Fasciola hepatica is a major public health and economic problem worldwide. Due to the lack of a successful vaccine and emerging resistance to the drug triclabendazole, alternative phytotherapeutic approaches are being investigated. This study investigated the in vitro anthelmintic activity of Lavender (Lavandula angustifolia) and carob (Ceratonia siliqua L.) essential oils (EOs) against F. hepatica. The in vitro study was based on an egg hatch assay (EHA), adult motility inhibition assays, DNA damage, reactive oxygen species (ROS) level along with several oxidative stress biomarkers including glutathione peroxidase (GSH), and glutathione-S-transferase (GST), superoxide dismutase (SOD) and malondialdehyde (MDA). To this end, different concentrations of L. angustifolia and C. siliqua EOs (1, 5, 10, 25 and 50 mg/mL) were used to assess anthelmintic effects on different life stages including egg, and adults of F. hepatica for 24 hrs. The results indicated that these EOs play a significant role as anthelminthics, and the effect was dependent on time and concentration. The in vitro treatment of F. hepatica worms with both L. angustifolia and C. siliqua EOs increased DNA damage, ROS production and induction of oxidative stress (decreased SOD, GST and GSH, and increased MDA), significantly compared to control. Therefore, it can be concluded that L. angustifolia and C. siliqua EOs have the potential to be used as novel agents for the control and treatment of F. hepatica infections. Further studies are required to investigate their pharmacological potential and effectiveness in vivo for the treatment of parasitic infections.


Asunto(s)
Antihelmínticos , Daño del ADN , Fasciola hepatica , Estrés Oxidativo , Plantas Medicinales , Animales , Estrés Oxidativo/efectos de los fármacos , Fasciola hepatica/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Antihelmínticos/farmacología , Plantas Medicinales/química , Biomarcadores , Especies Reactivas de Oxígeno/metabolismo , Aceites Volátiles/farmacología , Aceites Volátiles/química , Lavandula/química , Fascioliasis/tratamiento farmacológico , Fascioliasis/parasitología , Fascioliasis/veterinaria , Superóxido Dismutasa/metabolismo , Glutatión Transferasa/metabolismo , Glutatión Transferasa/genética , Estadios del Ciclo de Vida/efectos de los fármacos
20.
Molecules ; 29(11)2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38893380

RESUMEN

Breast cancer is a major health concern and the leading cause of death among women worldwide. Standard treatment often involves surgery, radiotherapy, and chemotherapy, but these come with side effects and limitations. Researchers are exploring natural compounds like baicalin and baicalein, derived from the Scutellaria baicalensis plant, as potential complementary therapies. This study investigated the effects of baicalin and baicalein on the cytotoxic, proapoptotic, and genotoxic activity of doxorubicin and docetaxel, commonly used chemotherapeutic drugs for breast cancer. The analysis included breast cancer cells (MCF-7) and human endothelial cells (HUVEC-ST), to assess potential effects on healthy tissues. We have found that baicalin and baicalein demonstrated cytotoxicity towards both cell lines, with more potent effects observed in baicalein. Both flavonoids, baicalin (167 µmol/L) and baicalein (95 µmol/L), synergistically enhanced the cytotoxic, proapoptotic, and genotoxic activity of doxorubicin and docetaxel in breast cancer cells. In comparison, their effects on endothelial cells were mixed and depended on concentration and time. The results suggest that baicalin and baicalein might be promising complementary agents to improve the efficacy of doxorubicin and docetaxel anticancer activity. However, further research is needed to validate their safety and efficacy in clinical trials.


Asunto(s)
Apoptosis , Neoplasias de la Mama , Docetaxel , Doxorrubicina , Flavanonas , Flavonoides , Humanos , Flavonoides/farmacología , Flavanonas/farmacología , Docetaxel/farmacología , Doxorrubicina/farmacología , Células MCF-7 , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Femenino , Daño del ADN/efectos de los fármacos , Sinergismo Farmacológico , Antineoplásicos/farmacología , Supervivencia Celular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...