Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 209
Filtrar
1.
Zhonghua Zhong Liu Za Zhi ; 44(9): 913-927, 2022 Sep 23.
Artículo en Chino | MEDLINE | ID: mdl-36164692

RESUMEN

As a newly emerged class of anticancer bioagents in the most precise and selectively targeted way, antibody-drug conjugate (ADC) combines the cancer-targeting abilities of monoclonal antibodies with the cytotoxicity potency of payload, delivering highly cytotoxic drug into tumors via 'targeted chemotherapy'. ADC has revolutionized the treatment landscape of human epidermal growth factor receptor 2 positive and triple negative subtypes in breast cancer. Three ADCs have been approved by U. S. Food and Drug Administration with breast cancer indications, including trastuzumab emtansine (T-DM1; also approved in China), trastuzumab deruxtecan (T-DXd, DS-8201) and sacituzumab govitecan (IMMU-132; also approved in China). Antibodies, cytotoxic drug, linker, and conjugation process are implicated in ADC profile, resulting in unique adverse drug reactions and toxicity heterogeneity within ADC class. For example, more attention should be paid to the management of thrombocytopenia, hepatotoxicity, and reductions in left ventricular ejection fraction (LVEF) in patients treated with trastuzumab emtansine; clinical physicians should pay attention to the risk of neutropenia, interstitial lung disease/pneumonitis, and reductions in LVEF when treated with trastuzumab deruxtecan; sacituzumab govitecan most frequently caused neutropenia, anemia and diarrhea requiring close monitor. ADC has generally favorable safety profiles, and dose modifications and/or symptomatic supporting treatment are effective in terms of toxicity management. This consensus aims at providing guidance for clinical oncologists of early detection, regular assessment, timely management and follow-up monitor of ADC-associated adverse reactions/events.


Asunto(s)
Antineoplásicos Inmunológicos , Neoplasias de la Mama , Citotoxinas , Inmunoconjugados , Neoplasias de la Mama Triple Negativas , Ado-Trastuzumab Emtansina/efectos adversos , Ado-Trastuzumab Emtansina/uso terapéutico , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/efectos adversos , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias de la Mama/terapia , Camptotecina/efectos adversos , Consenso , Citotoxinas/efectos adversos , Citotoxinas/uso terapéutico , Femenino , Humanos , Inmunoconjugados/efectos adversos , Inmunoconjugados/uso terapéutico , Neutropenia/etiología , Receptor ErbB-2/metabolismo , Trastuzumab/efectos adversos , Trastuzumab/uso terapéutico , Neoplasias de la Mama Triple Negativas/terapia , Función Ventricular Izquierda
2.
Mol Cell Biochem ; 477(1): 143-152, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34586566

RESUMEN

The heart is a very dynamic pumping organ working perpetually to maintain a constant blood supply to the whole body to transport oxygen and nutrients. Unfortunately, it is also subjected to various stresses based on physiological or pathological conditions, particularly more vulnerable to damages caused by oxidative stress. In this study, we investigate the molecular mechanism and contribution of IGF-IIRα in endoplasmic reticulum stress induction in the heart under doxorubicin-induced cardiotoxicity. Using in vitro H9c2 cells, in vivo transgenic rat cardiac tissues, siRNAs against CHOP, chemical ER chaperone PBA, and western blot experiments, we found that IGF-IIRα overexpression enhanced ER stress markers ATF4, ATF6, IRE1α, and PERK which were further aggravated by DOX treatment. This was accompanied by a significant perturbation in stress-associated MAPKs such as p38 and JNK. Interestingly, PARKIN, a stress responsive cellular protective mediator was significantly downregulated by IGF-IIRα concomitant with decreased expression of ER chaperone GRP78. Furthermore, ER stress-associated pro-apoptotic factor CHOP was increased considerably in a dose-dependent manner followed by elevated c-caspase-12 and c-caspase-3 activities. Conversely, treatment of H9c2 cells with chemical ER chaperone PBA or siRNA against CHOP abolished the IGF-IIRα-induced ER stress responses. Altogether, these findings suggested that IGF-IIRα contributes to ER stress induction and inhibits cellular stress coping proteins while increasing pro-apoptotic factors feeding into a cardio myocyte damage program that eventually paves the way to heart failure.


Asunto(s)
Estrés del Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Miocardio/metabolismo , Receptor IGF Tipo 2/metabolismo , Animales , Línea Celular , Citotoxinas/efectos adversos , Citotoxinas/farmacología , Doxorrubicina/efectos adversos , Doxorrubicina/farmacología , Retículo Endoplásmico/genética , Ratas , Ratas Transgénicas , Receptor IGF Tipo 2/genética
3.
Braz. J. Pharm. Sci. (Online) ; 58: e19542, 2022. graf
Artículo en Inglés | LILACS | ID: biblio-1384004

RESUMEN

Abstract The main aim of the study is to quantify the cytotoxic property of the Fucoidan extracted from the Turbinaria conoides using the MTT assay with the standard fucose. Fucoidan was extracted using the soaked water method and it was determined using the HPLC procedure the obtained Test sample Fucoidan extracted from the Turbinaria conoides and standard fucose was subjected to the cytotoxicity assay against the MCF7 Human breast cancer cell line, A549 lung cancer cell line, and L929 normal mouse fibroblast cell line. From the results it was found that the Test sample showed good IC50 value for MCF7 cell line then A549 with an increasing concentration 24 hours incubation at 37°C The IC50 for MCF7 was 115.21 µg/ml and A549 396.46µg/ml and the Fucoidan extract was checked for its cytotoxicity against the normal mouse fibroblast cell line L929, Fucoidan was found non-lethal to the L929 mouse fibroblast normal cell line. Standard fucose also gave a significant result towards MCF7 and against the L929. This indicates that the Fucoidan extracted from Tubinaria conoides shows better anticancer potential in it. Hence its application can be further extended in the pharmacological fields.


Asunto(s)
Técnicas In Vitro/instrumentación , Citotoxinas/efectos adversos , Células MCF-7 , Células A549 , Neoplasias de la Mama/patología , Línea Celular , Cromatografía Líquida de Alta Presión/métodos , Concentración 50 Inhibidora , Fibroblastos/clasificación , Fucosa/análogos & derivados , Neoplasias Pulmonares/patología
4.
Am J Physiol Lung Cell Mol Physiol ; 321(6): L1183-L1193, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34704847

RESUMEN

In chronic obstructive pulmonary disease (COPD), lung natural killer cells (NKs) lyse autologous lung epithelial cells in vitro, but underlying mechanisms and their relationship to epithelial cell apoptosis in vivo are undefined. Although this cytolytic capacity of lung NKs depends on priming by dendritic cells (DCs), whether priming correlates with DC maturation or is limited to a specific DC subset is also unknown. We recruited ever-smokers (≥10 pack-years; n = 96) undergoing clinically indicated lung resections. We analyzed lung NKs for cytotoxic molecule transcripts and for cytotoxicity, which we correlated with in situ detection of activated Caspase-3/7+ airway epithelial cells. To investigate DC priming, we measured lung DC expression of CCR2, CCR7, and CX3CR1 and cocultured peripheral blood NKs with autologous lung DCs, either matured using lipopolysaccharide (LPS) (nonobstructed smokers) or separated into conventional dendritic cell type-1 (cDC1) versus cDC type-2 (cDC2) (COPD). Lung NKs in COPD expressed more perforin (P < 0.02) and granzyme B (P < 0.03) transcripts; inhibiting perforin blocked in vitro killing by lung NKs. Cytotoxicity in vitro correlated significantly (Sr = 0.68, P = 0.0043) with numbers of apoptotic epithelial cells per airway. In nonobstructed smokers, LPS-induced maturation enhanced DC-mediated priming of blood NKs, reflected by greater epithelial cell death. Although CCR7 expression was greater in COPD in both cDC1 (P < 0.03) and cDC2 (P = 0.009), only lung cDC1 primed NK killing. Thus, rather than being intrinsic to those with COPD, NK priming is a capacity of human lung DCs that is inducible by recognition of bacterial (and possibly other) danger signals and restricted to the cDC1 subset.


Asunto(s)
Células Dendríticas/inmunología , Células Epiteliales/patología , Regulación de la Expresión Génica/efectos de los fármacos , Células Asesinas Naturales/inmunología , Pulmón/patología , Perforina/efectos adversos , Enfermedad Pulmonar Obstructiva Crónica/patología , Estudios de Casos y Controles , Citotoxinas/efectos adversos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Células Dendríticas/patología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Femenino , Granzimas/genética , Granzimas/metabolismo , Humanos , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/metabolismo , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Fumar/efectos adversos
5.
Nanomedicine ; 36: 102436, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34153528

RESUMEN

Platinum nanoparticles (PtNPs) have been investigated for their antioxidant abilities in a range of biological and other applications. The ability to reduce off-target cold atmospheric plasma (CAP) cytotoxicity would be useful in Plasma Medicine; however, little has been published to date about the ability of PtNPs to reduce or inhibit the effects of CAP. Here we investigate whether PtNPs can protect against CAP-induced cytotoxicity in cancerous and non-cancerous cell lines. PtNPs were shown to dramatically reduce intracellular reactive species (RONS) production in U-251 MG cells. However, RONS generation was unaffected by PtNPs in medium without cells. PtNPs protect against CAP induced mitochondrial membrane depolarization, but not cell membrane permeabilization which is a CAP-induced RONS-independent event. PtNPs act as potent intracellular scavengers of reactive species and can protect against CAP induced cytotoxicity. PtNPs, showing no significant biocorrosion, may be useful as a catalytic antioxidant for healthy tissue and for protecting against CAP-induced tissue damage.


Asunto(s)
Citotoxinas/efectos adversos , Nanopartículas del Metal , Estrés Oxidativo/efectos de los fármacos , Gases em Plasma/efectos adversos , Platino (Metal) , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral , Citotoxinas/farmacología , Células HEK293 , Humanos , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Gases em Plasma/farmacología , Platino (Metal)/química , Platino (Metal)/farmacología
6.
Anticancer Res ; 41(5): 2563-2568, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33952484

RESUMEN

BACKGROUND/AIM: The aim of this study was to evaluate the effect of drug-induced interstitial lung disease (DILD) on treatment outcomes by comparing the mortality of patients with DILD induced by different pharmacological types of anticancer drugs. PATIENTS AND METHODS: Japanese patients with lung cancer who had received chemotherapy at Fujita Health University Hospital were enrolled. The primary outcome was the short-term mortality rate from the administration of chemotherapy that might have caused DILD. RESULTS: Eleven, 16, and 20 patients with DILD were assigned to the kinase inhibitor (KI), immune-checkpoint inhibitor (ICI), and cytotoxic anticancer drug groups, respectively. The 90-day mortality rate after the DILD event in the group treated with cytotoxic anticancer drugs was significantly higher than in the KI and ICI groups. CONCLUSION: Patients with DILD induced by cytotoxic anticancer drugs have poorer prognoses than those with DILD induced by KIs or ICIs.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/mortalidad , Enfermedades Pulmonares Intersticiales/mortalidad , Neoplasias/mortalidad , Anciano , Anciano de 80 o más Años , Citotoxinas/administración & dosificación , Citotoxinas/efectos adversos , Quimioterapia , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Enfermedades Pulmonares Intersticiales/inducido químicamente , Enfermedades Pulmonares Intersticiales/tratamiento farmacológico , Enfermedades Pulmonares Intersticiales/patología , Masculino , Persona de Mediana Edad , Neoplasias/clasificación , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Pronóstico , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos
7.
Contact Dermatitis ; 85(3): 324-339, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34029376

RESUMEN

BACKGROUND: The number of people within the European population having at least one tattoo has increased notably, and with it the number of tattoo-associated clinical complications. Despite this, safety information and testing regarding tattoo inks remain limited. OBJECTIVE: To assess cytotoxicity and sensitization potential of 16 tattoo inks after intradermal injection into reconstructed human skin (RHS). METHODS: Commercially available tattoo inks were injected intradermally into RHS (reconstructed epidermis on a fibroblast-populated collagen hydrogel) using a permanent makeup device. RHS biopsies, tissue sections, and culture medium were assessed for cytotoxicity (thiazolyl blue tetrazolium bromide assay [MTT assay]), detrimental histological changes (haematoxylin and eosin staining), and the presence of inflammatory and sensitization cytokines (interleukin [IL]-1α, IL-8, IL-18; enzyme-linked immunosorbent assay). RESULTS: Varying degrees of reduced metabolic activity and histopathological cytotoxic effects were observed in RHS after ink injection. Five inks showed significantly reduced metabolic activity and enhanced sensitization potential compared with negative controls. DISCUSSION: Using the RHS model system, four tattoo inks were identified as highly cytotoxic and classified as potential sensitizers, suggesting that allergic contact dermatitis could emerge in individuals carrying these inks. These results indicate that an RHS-based assessment of cytotoxicity and sensitization potential by intradermal tattoo ink injection is a useful analytical tool to determine ink-induced deleterious effects.


Asunto(s)
Colorantes/efectos adversos , Citotoxinas/efectos adversos , Dermatitis Alérgica por Contacto/etiología , Tinta , Piel/patología , Tatuaje/efectos adversos , Citocinas/metabolismo , Fibroblastos , Humanos , Hidrogeles , Inyecciones Intradérmicas , Piel/inmunología , Piel/metabolismo
8.
Pancreas ; 50(4): 500-505, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33939660

RESUMEN

OBJECTIVES: Checkpoint inhibitors (CPIs) for low- and intermediate-grade neuroendocrine tumors (NETs) have been associated with limited efficacy; recent studies suggest CPIs may represent promising treatment for high-grade neuroendocrine neoplasms (NENs). METHODS: We examined 57 patients with NENs who were treated with CPIs to determine if NETs and neuroendocrine carcinomas (NECs) respond to immunotherapy. RESULTS: Patients with poorly differentiated NECs on CPI monotherapy had an objective response rate (ORR) of 0% and median progression-free survival (PFS) of 2.1 months (95% confidence interval [CI], 0.5-4.6). Patients with poorly differentiated NECs on dual CPI therapy had an ORR of 13% and PFS of 3.5 months (95% CI, 1.4-not reached [NR]). Patients with poorly differentiated NECs on CPI and cytotoxic therapy had an ORR of 36% with PFS of 4.2 months (95% CI, 1.6-NR). Well-differentiated grade 1 and 2 NETs on CPI monotherapy had an ORR of 25% with PFS NR. Well-differentiated grade 3 NETs had 0% ORR with a PFS of 2.9 months (95% CI, 1.4-4.2) on CPI monotherapy. CONCLUSIONS: Checkpoint inhibitor therapy shows limited activity in patients with NENs. Future studies should identify biomarkers that can help identify patients who are likely responders to immunotherapy.


Asunto(s)
Hospitales , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Tumores Neuroendocrinos/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Citotoxinas/administración & dosificación , Citotoxinas/efectos adversos , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/patología , Supervivencia sin Progresión , Tiroiditis/inducido químicamente , Resultado del Tratamiento , Adulto Joven
9.
FASEB J ; 35(6): e21640, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33991130

RESUMEN

Certain species of pathogenic bacteria damage tissues by secreting cholesterol-dependent cytolysins, which form pores in the plasma membranes of animal cells. However, reducing cholesterol protects cells against these cytolysins. As the first committed step of cholesterol biosynthesis is catalyzed by squalene synthase, we explored whether inhibiting this enzyme protected cells against cholesterol-dependent cytolysins. We first synthesized 22 different nitrogen-containing bisphosphonate molecules that were designed to inhibit squalene synthase. Squalene synthase inhibition was quantified using a cell-free enzyme assay, and validated by computer modeling of bisphosphonate molecules binding to squalene synthase. The bisphosphonates were then screened for their ability to protect HeLa cells against the damage caused by the cholesterol-dependent cytolysin, pyolysin. The most effective bisphosphonate reduced pyolysin-induced leakage of lactate dehydrogenase into cell supernatants by >80%, and reduced pyolysin-induced cytolysis from >75% to <25%. In addition, this bisphosphonate reduced pyolysin-induced leakage of potassium from cells, limited changes in the cytoskeleton, prevented mitogen-activated protein kinases cell stress responses, and reduced cellular cholesterol. The bisphosphonate also protected cells against another cholesterol-dependent cytolysin, streptolysin O, and protected lung epithelial cells and primary dermal fibroblasts against cytolysis. Our findings imply that treatment with bisphosphonates that inhibit squalene synthase might help protect tissues against pathogenic bacteria that secrete cholesterol-dependent cytolysins.


Asunto(s)
Colesterol/metabolismo , Citotoxinas/efectos adversos , Difosfonatos/farmacología , Inhibidores Enzimáticos/farmacología , Farnesil Difosfato Farnesil Transferasa/antagonistas & inhibidores , Fibroblastos/citología , Sustancias Protectoras/farmacología , Células A549 , Proteínas Bacterianas/efectos adversos , Toxinas Bacterianas/efectos adversos , Proliferación Celular , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Células HeLa , Proteínas Hemolisinas/efectos adversos , Humanos , Estreptolisinas/efectos adversos
10.
Toxicol Ind Health ; 37(5): 270-279, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33856234

RESUMEN

The organochlorine insecticide dichlorodiphenyltrichloroethane (DDT) and heavy metal cadmium (Cd) are widespread environmental pollutants. They are persistent in the environment and can accumulate in organisms. Although the individual toxicity of DDT and Cd has been well documented, their combined toxicity is still not clear. Since liver is their common target, in this study, the individual and combined toxicity of DDT and Cd in human liver carcinoma HepG2 and human normal liver THLE-3 cell lines were investigated. The results showed that DDT and Cd inhibited the viability of HepG2 and THLE-3 cells dose-dependently and altered lysosomal morphology and function. Intracellular reactive oxygen species and lipid peroxidation levels were induced by DDT and Cd treatment. The combined cytotoxicity of DDT and Cd was greater than their individual cytotoxicity, and the interaction between Cd and DDT was additive on the inhibition of cell viability and lysosomal function of HepG2 cells. The interaction was antagonistic on the inhibition of cell viability of THLE-3 cells. These results may facilitate the evaluation of the cumulative risk of pesticides and heavy metal residues in the environment.


Asunto(s)
Cadmio/toxicidad , Supervivencia Celular/efectos de los fármacos , Citotoxinas/efectos adversos , DDT/toxicidad , Contaminantes Ambientales/toxicidad , Células Hep G2/efectos de los fármacos , Insecticidas/toxicidad , Metales Pesados/toxicidad , Células Cultivadas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Estrés Oxidativo/efectos de los fármacos
11.
J Appl Toxicol ; 41(10): 1660-1672, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33624853

RESUMEN

Nodularin (NOD) is a cyclic peptide released by bloom-forming toxic cyanobacteria Nodularia spumigena commonly occurring in brackish waters throughout the world. Although its hepatotoxic effects are well known, other negative effects of NOD have not yet been completely elucidated. The present study aims were to evaluate and compare the cytotoxic and immunotoxic effects of the toxin on primary leukocytes (from head kidney [HK]) and stable fish leukocytes (carp leucocyte cell line [CLC] cells). The cells were incubated with the cyanotoxin at concentrations of 0.001, 0.01, 0.05, or 0.1 µg/ml. After 24 h of exposure, the concentrations ≥0.05 µg/ml of toxin resulted in cytotoxicity in the primary cells, while in CLC cells, the toxic effect was obtained only with the highest concentration. Similarly, depending on the concentration, exposure to NOD causes a significant inhibition of chemotaxis of the phagocytic abilities of primary leukocytes and a significant reduction in the proliferation of lymphocytes isolated from the HKs. Moreover, CLC cells and HK leukocytes incubated with this toxin at all the mentioned concentrations showed an increased production of reactive oxygen and nitrogen species. NOD also evidently influenced the expression of genes of cytokine TNF-α and IL-10 and, to a minor extent, IL-1ß and TGF-ß. Notably, the observed changes in the mRNA levels of cytokines in NOD-exposed cells were evident, but not clearly dose-dependent. Interestingly, NOD did not affect the production and release of IL-1ß of the CLC cells. This study provides evidence that NOD may exert cytotoxicity and immune-toxicity effects depending on cell type and toxin concentration.


Asunto(s)
Toxinas Bacterianas/toxicidad , Carpas/crecimiento & desarrollo , Células Cultivadas/efectos de los fármacos , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Péptidos Cíclicos/toxicidad , Animales , Citotoxinas/efectos adversos , Leucocitos/inmunología , Nodularia/química
12.
J Biochem Mol Toxicol ; 35(4): e22699, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33398916

RESUMEN

Statins are a group of hydroxymethylglutaryl coenzyme A reductase inhibitors that are used in the treatment of cardiovascular diseases. However, statins have been found to be cytotoxic, and many unexpected side effects have been reported in clinical applications. The susceptibilities of different cell lines toward statins are diverse, and the mechanisms of cytotoxicity remain unknown. Therefore, the present study aimed to investigate differences in the susceptibility to and mechanisms of statin-induced cytotoxicity in two cell lines, HT-29 and A549, using a high content screening-based multiparametric toxicity assay panel. We found that the two cell types exhibited differing susceptibilities to the cytotoxic effects of the different statins. Additionally, the cytotoxicity was inconsistent between different statins in the two cell lines. Four statins with strong cytotoxicity decreased the viability of HT-29 cells via the mitochondrial pathway, as evidenced by decreased mitochondrial membrane potential, and elevated mitochondrial mass, calcium release and cell apoptosis, and reactive oxygen species. In contrast, these four statins only induced a decrease in the mitochondrial membrane potential in A549 cells. The above results provide an objective reason for future evaluations of cytotoxic differences in cell types and the underlying mechanisms of cytotoxicity in different statins, and provide a good scientific basis for further research on countermeasures against statin-induced cell injuries.


Asunto(s)
Apoptosis/efectos de los fármacos , Citotoxinas/efectos adversos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/metabolismo , Células A549 , Supervivencia Celular/efectos de los fármacos , Citotoxinas/farmacología , Células HT29 , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Mitocondrias/patología
13.
J Biochem Mol Toxicol ; 35(4): e22712, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33484013

RESUMEN

Asiatic acid (AA) is a triterpene with promising pharmacological activity. In the present study, in vitro and in vivo assays were conducted to understand the effect of AA on cell proliferation and genomic instability. AA was cytotoxic to human tumor cell lines (M059J, HeLa, and MCF-7), with IC50 values ranging from 13.91 to 111.72 µM. In the case of M059J, AA exhibited selective cytotoxicity after 48 h of treatment (IC50 = 24 µM), decreasing the percentage of cells in the G0/G1 phase, increasing the percentage of cells in the S phase, and inducing apoptosis. A significant increase in chromosomal damage was observed in V79 cell cultures treated with AA (40 µM), revealing genotoxic activity. In contrast, low concentrations (5, 10, and 20 µM) of AA significantly reduced the frequencies of micronuclei induced by the mutagens doxorubicin (DXR), methyl methanesulfonate, and hydrogen peroxide. A reduction of DXR-induced intracellular free radicals was found in V79 cells treated with AA (10 µM). The antigenotoxic effect of AA (30 mg/kg) was also observed against DXR-induced chromosomal damage in Swiss mice. Significant reductions in p53 levels were verified in the liver tissue of these animals. Taken together, the data indicate that AA exerted antiproliferative activity in M059J tumor cells, which is probably related to the induction of DNA damage, leading to cell cycle arrest and apoptosis. Additionally, low concentrations of AA exhibited antigenotoxic effects and its antioxidant activity may be responsible, at least in part, for chemoprevention.


Asunto(s)
Antioxidantes/farmacología , Ciclo Celular/efectos de los fármacos , Daño del ADN , Triterpenos Pentacíclicos/farmacología , Animales , Cricetulus , Citotoxinas/efectos adversos , Citotoxinas/farmacología , Doxorrubicina/efectos adversos , Doxorrubicina/farmacología , Células HeLa , Humanos , Células MCF-7 , Masculino , Ratones
14.
J Biochem Mol Toxicol ; 35(3): e22683, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33325091

RESUMEN

Cardiotoxicity is a critical concern in the use of several cytotoxic drugs. Induction of apoptosis, inflammation, and autophagy following dysregulation of the PI3K/Akt signaling pathway contributes to the cardiac damage induced by these drugs. Several natural compounds (NCs), including ferulic acid, gingerol, salvianolic acid B, paeonol, apigenin, calycosin, rutin, neferine, higenamine, vincristine, micheliolide, astragaloside IV, and astragalus polysaccharide, have been reported to suppress cytotoxic drug-induced cardiac injury. This article reviews these NCs that have been reported to have a protective effect against cytotoxic drug-induced cardiotoxicity through regulation of the PI3K/Akt signaling pathway.


Asunto(s)
Cardiotoxicidad , Citotoxinas/efectos adversos , Fosfatidilinositol 3-Quinasas/metabolismo , Fitoquímicos/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxicidad/metabolismo , Citotoxinas/uso terapéutico , Humanos
15.
Respir Med ; 172: 106131, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32911136

RESUMEN

BACKGROUND: High-mobility group box 1 (HMGB1) is a pro-inflammatory protein, that is associated with tumorigenesis, interstitial lung disease (ILD), and acute lung injury. Chemotherapy-induced lung injury is a common and serious adverse event in patients with lung cancer and ILD, but its pathogenesis and predictive biomarkers are not known. This study aimed to investigate the predictive potential of serum HMGB1 levels for cytotoxic chemotherapy-induced lung injury in these patients. METHODS: From 743 patients with advanced lung cancer, we enrolled 83 consecutive patients with ILD and background-matched 83 patients without ILD. Additionally, 83 healthy subjects were included. After measuring baseline levels of serum HMGB1 in three groups, we evaluated the predictive values of baseline HMGB1 levels for cytotoxic chemotherapy-induced lung injury in patients with lung cancer and ILD. RESULTS: Higher levels of serum HMGB1 were independently associated with higher tumor burden, as assessed by total tumor size, and the presence of ILD. Twenty-five (30.1%) of patients with lung cancer and ILD experienced cytotoxic chemotherapy-induced lung injury within one year. Univariate Cox proportional hazards model showed that higher levels of HMGB1 and higher tumor burden were associated with disease onset. Moreover, multivariate analysis revealed that only HMGB1 was independently associated with this severe complication in patients with lung cancer and ILD. CONCLUSIONS: HMGB1 is a potential predictive blood biomarker for cytotoxic chemotherapy-induced lung injury in patients with lung cancer and ILD. This study also suggests a potential pathogenesis of this serious adverse event that tumor- and ILD-derived HMGB1 accelerates lung injury.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/diagnóstico , Antineoplásicos/efectos adversos , Citotoxinas/efectos adversos , Proteínas del Grupo de Alta Movilidad/sangre , Enfermedades Pulmonares Intersticiales/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Lesión Pulmonar Aguda/etiología , Anciano , Biomarcadores/sangre , Femenino , Humanos , Enfermedades Pulmonares Intersticiales/complicaciones , Neoplasias Pulmonares/complicaciones , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas
16.
Nanotechnology ; 31(46): 465102, 2020 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-32857735

RESUMEN

The biological effects of nanoparticles are of great importance for the in-depth understanding of safety issues in biomedical applications. Induction of autophagy is a cellular response after nanoparticle exposure. Bismuth sulfide nanoparticles (Bi2S3 NPs) are often used as a CT contrast agent because of their excellent photoelectric conversion ability. Yet there has been no previous detailed study other than a cell toxicity assessment. In this study, three types of Bi2S3 NPs with different shapes (Bi2S3 nano rods (BSNR), hollow microsphere Bi2S3 NPs (BSHS) and urchin-like hollow microsphere Bi2S3 NPs (ULBSHS)) were used to evaluatecytotoxicity, autophagy induction, cell migration and invasion in human hepatocellular carcinoma cells (HepG2). Results showed that all three Bi2S3 NPs lead to blockage in autophagic flux, causing p62 protein accumulation. The cell death caused by these Bi2S3 NPs is proved to be autophagy related, rather than related to apoptosis. Moreover, Bi2S3 NPs can reduce the migration and invasion in HepG2 cells in an autophagy-dependent manner. ULBSHS is the most cytotoxic among three Bi2S3 NPs and has the best tumor metastasis suppression. These results demonstrated that, even with relatively low toxicity of Bi2S3 NPs, autophagy blockage may still substantially influence cell fate and thus significantly impact their biomedical applications, and that surface topography is a key factor regulating their biological response.


Asunto(s)
Autofagia/efectos de los fármacos , Bismuto/efectos adversos , Movimiento Celular/efectos de los fármacos , Citotoxinas/efectos adversos , Nanopartículas/efectos adversos , Sulfuros/efectos adversos , Bismuto/química , Bismuto/toxicidad , Citotoxinas/química , Citotoxinas/toxicidad , Células Hep G2 , Humanos , Nanopartículas/química , Nanopartículas/toxicidad , Sulfuros/química , Sulfuros/toxicidad
17.
Eur J Haematol ; 105(4): 476-483, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32544294

RESUMEN

OBJECTIVES: We sought to characterise the outcomes of patients with haematological malignancy and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in hospital in our regional network of 7 hospitals. METHODS: Consecutive hospitalised patients with haematological malignancy and SARS-CoV-2 infection were identified from 01/03/2020 to 06/05/2020. Outcomes were categorised as death, resolved or ongoing. The primary outcome was preliminary case fatality rate (pCFR), defined as the number of cases resulting in death as a proportion of all diagnosed cases. Analysis was primarily descriptive. RESULTS: 66 Patients were included, overall pCFR was 51.5%. Patients ≥ 70 years accounted for the majority of hospitalised cases (42, 63%) and fatalities (25, 74%). Mortality was similar between females (52%) and males (51%). Immunosuppressive or cytotoxic treatment within 3 months of the diagnosis of SARS-CoV-2 infection was associated with a significantly higher pCFR of 70%, compared with 28% in those not on active treatment (P = .0013, 2 proportions z test). CONCLUSIONS: Mortality rates in patients with haematological malignancy and SARS-CoV-2 infection in hospital are high supporting measures to minimise the risk of infection in this population.


Asunto(s)
COVID-19/complicaciones , Neoplasias Hematológicas/complicaciones , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , COVID-19/mortalidad , COVID-19/prevención & control , Citotoxinas/efectos adversos , Femenino , Neoplasias Hematológicas/terapia , Hospitalización , Humanos , Terapia de Inmunosupresión/efectos adversos , Masculino , Persona de Mediana Edad , Pandemias , Estudios Prospectivos , SARS-CoV-2 , Reino Unido/epidemiología
18.
Am Soc Clin Oncol Educ Book ; 40: 485-500, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32421446

RESUMEN

Over the past 2 decades, rapid advancement in systemic anticancer therapeutics has led to astounding improvement in survival rates of patients with cancer. However, this celebrated progress has brought with it an evolving spectrum of drug toxicities that limit their prodigious capabilities. Cutaneous adverse events are of the most frequent of these toxicities, with substantial impact on quality of life and commonly resulting in dose reduction or change in therapy. Thus, familiarity with the array of dermatologic manifestations caused by these drugs is prudent for patient treatment. As such, the advent of dedicated oncodermatologists, and their introduction into multidisciplinary cancer care, has been crucial in optimizing treatment through therapeutic achievement and overall well-being. This review will address the epidemiology, clinical presentations, and management strategies of the major dermatologic adverse events of systemic anticancer agents, including cytotoxic chemotherapy, targeted therapy, and immunotherapy.


Asunto(s)
Antineoplásicos/efectos adversos , Citotoxinas/efectos adversos , Inmunoterapia/efectos adversos , Terapia Molecular Dirigida/efectos adversos , Neoplasias/terapia , Piel/efectos de los fármacos , Humanos
19.
J Inorg Biochem ; 208: 111082, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32413634

RESUMEN

Several biologically active bivalent Pd and Pt complexes with two structurally similar cyanoxime ligands abbreviated as H(DECO): 2-oximino-2-cyano-N,N'-diethylacetamide, and H(PyrCO): 2-oximino-2-cyan-N-pyrrolidine acetamide were synthesized and characterized using spectroscopic methods, thermal analysis and X-ray crystallography. Structures revealed planar cis-geometry of studied complexes. Freshly obtained Pt(DECO)2, Pd(DECO)2, Pt(PyrCO)2 and Pd(PyrCO)2 complexes were used in for in vitro cytotoxicity assays using two different etiology human cancer cell lines HeLa and WiDr cells. Investigated compounds showed cytotoxicity levels at or above cisplatin. Pt(DECO)2 was also tested in vivo in healthy C57BL/6 mice. The complex was administered at three different dosage (0, 7.5, 15 mg/kg, i.p. once/week), over a total period of 8 weeks. No changes were observed in the animal weight in the treated mice compared to the control dextrose-treated group. The levels of erythrocytes, leukocytes, and hemoglobin were within the normal level suggesting low myelotoxicity. Negligible cardiotoxicity was observed from the histological evaluation of the hearts from the treated animals. Results from the tail nerve conduction velocity (NCV) and nerve histomorphometry suggested no impact of Pt(DECO)2 on peripheral nerves. The complex, however, induced certain hepatotoxicity and lead to the elevation of IL-6, a pro-inflammatory cytokine. Overall, Pt(DECO)2 showed minimal in vivo toxicity, thus presenting a promising candidate for future testing in animal models of cancer.


Asunto(s)
Complejos de Coordinación , Citotoxinas , Neoplasias/tratamiento farmacológico , Paladio , Platino (Metal) , Animales , Complejos de Coordinación/efectos adversos , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Citotoxinas/efectos adversos , Citotoxinas/síntesis química , Citotoxinas/química , Citotoxinas/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Células HeLa , Humanos , Masculino , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Paladio/efectos adversos , Paladio/química , Paladio/farmacología , Platino (Metal)/efectos adversos , Platino (Metal)/química , Platino (Metal)/farmacología
20.
J Appl Toxicol ; 40(7): 918-930, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32080871

RESUMEN

Ultrasmall superparamagnetic iron oxide nanoparticles (USPION) possess reactive surfaces, are metabolized and exhibit unique magnetic properties. These properties are desirable for designing novel theranostic biomedical products; however, toxicity mechanisms of USPION are not completely elucidated. The goal of this study was to investigate cell interactions (uptake and cytotoxicity) of USPION using human coronary artery endothelial cells as a vascular cell model. Polyvinylpirrolidone-coated USPION were characterized: average diameter 17 nm (transmission electron microscopy [TEM]), average hydrodynamic diameter 44 nm (dynamic light scattering) and zeta potential -38.75 mV. Cells were exposed to 0 (control), 25, 50, 100 or 200 µg/mL USPION. Concentration- and time-dependent cytotoxicity were observed after 3-6 hours through 24 hours of exposure using Alamar Blue and Real-Time Cell Electronic Sensing assays. Cell uptake was evaluated by imaging using live-dead confocal microscopy, actin and nuclear fluorescent staining, and TEM. Phase-contrast, confocal microscopy, and TEM imaging showed significant USPION internalization as early as 3 hours after exposure to 25 µg/mL. TEM imaging demonstrated particle internalization in secondary lysosomes with perinuclear localization. Three orthogonal assays were conducted to assess apoptosis. TUNEL staining demonstrated a marked increase in fragmented DNA, a response pathognomonic of apoptosis, after a 4-hour exposure. Cells subjected to agarose gel electrophoresis exhibited degraded DNA 3 hours after exposure. Caspase-3/7 activity increased after a 3-hour exposure. USPION uptake resulted in cytotoxicity involving apoptosis and these results contribute to further mechanistic understanding of the USPION toxicity in vitro in cardiovascular endothelial cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Transporte Biológico/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Vasos Coronarios/efectos de los fármacos , Citotoxinas/efectos adversos , Células Endoteliales/efectos de los fármacos , Nanopartículas Magnéticas de Óxido de Hierro/toxicidad , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...