Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.674
Filtrar
1.
Toxicol Appl Pharmacol ; 438: 115830, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34933053

RESUMEN

Dibenzo[def,p]chrysene (DBC) is an environmental polycyclic aromatic hydrocarbon (PAH) that causes tumors in mice and has been classified as a probable human carcinogen by the International Agency for Research on Cancer. Animal toxicity studies often utilize higher doses than are found in relevant human exposures. Additionally, like many PAHs, DBC requires metabolic bioactivation to form the ultimate toxicant, and species differences in DBC and DBC metabolite metabolism have been observed. To understand the implications of dose and species differences, a physiologically based pharmacokinetic model (PBPK) for DBC and major metabolites was developed in mice and humans. Metabolism parameters used in the model were obtained from experimental in vitro metabolism assays using mice and human hepatic microsomes. PBPK model simulations were evaluated against mice dosed with 15 mg/kg DBC by oral gavage and human volunteers orally microdosed with 29 ng of DBC. DBC and its primary metabolite DBC-11,12-diol were measured in blood of mice and humans, while in urine, the majority of DBC metabolites were obeserved as conjugated DBC-11,12-diol, conjugated DBC tetrols, and unconjugated DBC tetrols. The PBPK model was able to predict the time course concentrations of DBC, DBC-11,12-diol, and other DBC metabolites in blood and urine of human volunteers and mice with reasonable accuracy. Agreement between model simulations and measured pharmacokinetic data in mice and human studies demonstrate the success and versatility of our model for interspecies extrapolation and applicability for different doses. Furthermore, our simulations show that internal dose metrics used for risk assessment do not necessarily scale allometrically, and that PBPK modeling provides a reliable approach to appropriately account for interspecies differences in metabolism and physiology.


Asunto(s)
Crisenos/administración & dosificación , Crisenos/farmacocinética , Cistina/análogos & derivados , Animales , Carcinógenos/administración & dosificación , Carcinógenos/farmacocinética , Cistina/administración & dosificación , Cistina/farmacocinética , Femenino , Humanos , Masculino , Ratones , Modelos Biológicos , Neoplasias/inducido químicamente
2.
Toxins (Basel) ; 13(11)2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34822609

RESUMEN

It is important to note that 2-Amino-3-methylimidazole[4,5-f]quinoline (IQ) is one of the most common heterocyclic amines (HCAs), which is a class of mutagenic/carcinogenic harmful compounds mainly found in high-protein thermal processed foods and contaminated environments. However, the pre-carcinogenic toxicity of IQ to the liver and its mechanism are poorly understood, further research is needed. In light of this, we exposed zebrafish to IQ (0, 8, 80, and 800 ng/mL) for 35 days, followed by comprehensive experimental studies. Histopathological and ultrastructural analysis showed that hepatocytes were damaged. TUNEL results showed that IQ induced apoptosis of liver cells, the expression of apoptosis factor gene was significantly increased, and the expression of Bcl-2 protein was significantly decreased. In addition, upregulated expression of the 78-kDa glucose-regulated protein (GRP78) and C/EBP homologous protein (CHOP) and endoplasmic reticulum stress (ERS)-related factors transcription levels were elevated obviously, suggesting that IQ induced ERS. Decreased protein expression of autophagy-related 5 (Atg5)-Atg12, Beclin1, and LC3-II, increased protein expression of p62, and autophagy-related factors transcription levels were significantly decreased, suggesting that IQ inhibited autophagy. Overall, our research showed that the potential harm of IQ to the liver before the occurrence of liver cancer was related to ERS and its mediated autophagy and apoptosis pathways.


Asunto(s)
Carcinógenos/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Quinolinas/toxicidad , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Carcinógenos/administración & dosificación , Enfermedad Hepática Inducida por Sustancias y Drogas/fisiopatología , Relación Dosis-Respuesta a Droga , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Etiquetado Corte-Fin in Situ , Quinolinas/administración & dosificación , Pez Cebra
3.
Eur J Pharmacol ; 913: 174626, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34774852

RESUMEN

Disruption in the nerve-tumor interaction is now considered as a possible anticancer strategy for treating various cancer types, particularly colorectal cancer. However, the underlying mechanisms are not still fully understood. Therefore, the present study aimed to evaluate the effects of sympathetic and parasympathetic denervation on the inhibition of colorectal cancer progression in early and late phases and assess the involvement of nerve growth factor in denervation mediated anticancer effects. One-hundred and fifty male Wistar rats were assigned into 15 groups. Seven groups comprising the control group, 1,2-dimethylhydrazine (DMH) group, sympathetic denervation group (celiac-mesenteric ganglionectomy and guanethidine sulphate administration), parasympathetic denervation group (vagotomy and atropine administration), and combination group were used in the early-stage protocol. For the late-stage protocol, eight groups comprising the control, DMH, surgical and pharmacological sympathetic and parasympathetic denervation groups, combination group, and 5-flourouracil group were considered. After 8 weeks, sympathetic and parasympathetic denervation significantly reduced ACF numbers in rats receiving DMH. On the other hand, in the late stages, parasympathetic but not sympathetic denervation resulted in significant reductions in tumor incidence, tumor volume and weight, cell proliferation (indicated by reduced immunostaining of PCNA and ki-67), and angiogenesis (indicated by reduced immunostaining of CD31 and VEGF expression levels), and downregulated NGF, ß2 adrenergic, and M3 receptors. It can be concluded that parasympathetic denervation may be of high importance in colon carcinogenesis and suggested as a possible therapeutic modality in late stages of colorectal cancer.


Asunto(s)
Atropina/administración & dosificación , Neoplasias Colorrectales/cirugía , Neoplasias Experimentales/cirugía , Vagotomía , 1,2-Dimetilhidrazina/administración & dosificación , 1,2-Dimetilhidrazina/toxicidad , Animales , Carcinogénesis/inducido químicamente , Carcinógenos/administración & dosificación , Carcinógenos/toxicidad , Colon/inervación , Colon/patología , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Ganglios Simpáticos/efectos de los fármacos , Ganglios Simpáticos/cirugía , Ganglionectomía , Guanetidina/administración & dosificación , Humanos , Masculino , Mesenterio/inervación , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/patología , Sistema Nervioso Parasimpático/efectos de los fármacos , Sistema Nervioso Parasimpático/cirugía , Ratas , Ratas Wistar
4.
Toxicology ; 463: 152987, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34648870

RESUMEN

1,3-Butadiene (BD) exposure is known to cause numerous adverse health effects, including cancer, in animals and humans. BD is metabolized to reactive epoxide intermediates, which are genotoxic, but it is not well know what other effects BD has on cellular metabolism. We examined the effects of exposure to BD on the mouse lung metabolome in the genetically heterogeneous collaborative cross outbred mouse model. Mice were exposed to 3 concentra-tions of BD for 10 days (2, 20, and 200 ppm), and lung tissues were analyzed using high-resolution mass spectrometry-based metabolomics. As compared to controls (0 ppm BD), BD had extensive effects on lung metabolism at all concentrations of exposure, including the lowest concentration of 2 ppm, as reflected by reprogramming of multiple metabolic pathways. Metabolites participating in glycolysis and the tricarboxylic acid cycle were elevated, with 8 out of 10 metabolites demonstrating a 2 to 8-fold increase, including the oncometabolite fumarate. Fatty acid levels, sphingosine, and sphinganine were decreased (2 to 8-fold), and fatty acyl-CoAs were significantly increased (16 to 31-fold), suggesting adjustments in lipid metabolism. Furthermore, metabolites involved in basic amino acid metabolism, steroid hormone metabolism, and nucleic acid metabolism were significantly altered. Overall, these changes mirror the metabolic alterations found in lung cancer cells, suggesting that very low doses of BD induce metabolic adaptations that may prevent or promote adverse health effects such as tumor formation.


Asunto(s)
Butadienos/toxicidad , Neoplasias Pulmonares/patología , Pulmón/patología , Metabolómica , Animales , Butadienos/administración & dosificación , Butadienos/metabolismo , Carcinógenos/administración & dosificación , Carcinógenos/metabolismo , Carcinógenos/toxicidad , Ratones de Colaboración Cruzada , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Pulmón/metabolismo , Neoplasias Pulmonares/metabolismo , Espectrometría de Masas , Metaboloma , Ratones , Fenotipo
5.
Sci Rep ; 11(1): 20672, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34667240

RESUMEN

Peri-implantitis may result in the loss of dental implants. Cold atmospheric pressure plasma (CAP) was suggested to promote re-osseointegration, decrease antimicrobial burden, and support wound healing. However, the long-term risk assessment of CAP treatment in the oral cavity has not been addressed. Treatment with two different CAP devices was compared against UV radiation, carcinogen administration, and untreated conditions over 12 months. Histological analysis of 406 animals revealed that repeated CAP exposure did not foster non-invasive lesions or squamous cell carcinoma (SCCs). Carcinogen administration promoted non-invasive lesions and SCCs. Molecular analysis by a qPCR screening of 144 transcripts revealed distinct inflammatory profiles associated with each treatment regimen. Interestingly, CAP treatment of carcinogen-challenged mucosa did not promote but instead left unchanged or reduced the proportion of non-invasive lesions and SCC formation. In conclusion, repeated CAP exposure of murine oral mucosa was well tolerated, and carcinogenic effects did not occur, motivating CAP applications in patients for dental and implant treatments in the future.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Carcinógenos/administración & dosificación , Mucosa Bucal/efectos de los fármacos , Gases em Plasma/administración & dosificación , Animales , Antibacterianos/farmacología , Presión Atmosférica , Implantes Dentales/efectos adversos , Inflamación/inducido químicamente , Masculino , Ratones , Oseointegración/efectos de los fármacos , Periimplantitis/inducido químicamente , Propiedades de Superficie/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos
6.
Neurotoxicology ; 87: 219-230, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34687775

RESUMEN

Galaxolide and tonalide are well-known polycyclic musks whose intensive use without limitations in numerous cleaning, hygiene, and personal care products has resulted in widespread direct human exposure via absorption, inhalation, and oral ingestion. Latest data shows that long-term, low-dose exposure to toxic chemicals can induce unpredictable harmful effects in a variety of living systems, however, interactions between synthetic musks and brain tumours remain largely unexplored. Glioblastoma (GB) accounts for nearly half of all tumours of the central nervous system and is characterized by very poor prognosis. The aims of this study were (1) to investigate the potential effect of long-term (20-generation) single and combined application of galaxolide and tonalide at sub-lethal doses (5-2.5 u M) on the angiogenesis, invasion, and migration of human U87 cells or tumour spheroids, and (2) to explore the underlying molecular mechanisms. Random amplified polymorphic DNA assays revealed significant DNA damage and increased total mutation load in galaxolide- and/or tonalide-treated U87 cells. In those same groups, we also detected remarkable tumour spheroid invasion and up-regulation of both HIF1-α/VEGF/MMP9 and IL6/JAK2/STAT3 signals, known to have important roles in hypoxia-related angiogenesis and/or proliferation. Prolonged musk treatment further altered angio-miRNA expression in a manner consistent with poor prognosis in GB. We also detected significant over-expression of the genes Slug, Snail, ZEB1, and Vimentin, which are biomarkers of epithelial to mesenchymal transition. In addition, matrigel, transwell, and wound healing assays clearly showed that long-term sub-lethal exposure to galaxolide and/or tonalide induced invasion and migration proposing a high metastatic potential. Our results suggest that assessing expression of HIF-1a, VEGF, STAT3, and the miR-17-92 cluster in biopsy samples of GB patients who have a history of possible long-term exposure to galaxolide or tonalide could be beneficial for deciding a therapy regime. Additionally, we recommend that extensively-used hygiene and cleaning materials be selected from synthetic musk-free products, especially when used in palliative care processes for GB patients.


Asunto(s)
Benzopiranos/toxicidad , Carcinógenos/toxicidad , Glioblastoma/inducido químicamente , Tetrahidronaftalenos/toxicidad , Benzopiranos/administración & dosificación , Carcinógenos/administración & dosificación , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/efectos de los fármacos , Glioblastoma/patología , Humanos , Esferoides Celulares/efectos de los fármacos , Tetrahidronaftalenos/administración & dosificación
7.
Toxicology ; 462: 152936, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34509578

RESUMEN

Non-human primates (NHPs) have played a vital role in fundamental, pre-clinical, and translational studies because of their high physiological and genetic similarity to humans. Here, we report a method to isolate primary hepatocytes from the livers of rhesus macaques (Macaca mulatta) after in situ whole liver perfusion. Isolated primary macaque hepatocytes (PMHs) were treated with various compounds known to have different pathways of genotoxicity/carcinogenicity and the resulting DNA damage was evaluated using the high-throughput CometChip assay. The comet data were quantified using benchmark dose (BMD) modeling and the BMD50 values for treatments of PMHs were compared with those generated from primary human hepatocytes (PHHs) in our previous study (Seo et al. Arch Toxicol 2020, 2207-2224). The results showed that despite varying CYP450 enzyme activities, PMHs had the same sensitivity and specificity as PHHs in detecting four indirect-acting (i.e., requiring metabolic activation) and seven direct-acting genotoxicants/carcinogens, as well as five non-carcinogens that are negative or equivocal for genotoxicity in vivo. The BMD50 estimates and their confidence intervals revealed species differences for DNA damage potency, especially for direct-acting compounds. The present study provides a practical method for maximizing the use of animal tissues by isolating primary hepatocytes from NHPs. Our data support the use of PMHs as a reliable surrogate of PHHs for evaluating the genotoxic hazards of chemical substances for humans.


Asunto(s)
Carcinógenos/toxicidad , Daño del ADN/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Mutágenos/toxicidad , Animales , Benchmarking , Carcinógenos/administración & dosificación , Sistema Enzimático del Citocromo P-450/metabolismo , Relación Dosis-Respuesta a Droga , Hepatocitos/enzimología , Hepatocitos/patología , Ensayos Analíticos de Alto Rendimiento , Humanos , Macaca mulatta , Masculino , Mutágenos/administración & dosificación , Reproducibilidad de los Resultados , Especificidad de la Especie
8.
Biomed Pharmacother ; 142: 112081, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34463271

RESUMEN

Previous reports demonstrated that aristolochic acids (AAs) exposure-induced nephrotoxicity, mutations, and tumorigenesis are mainly due to aristolochic acid I (AAI). Notably, the chemical structure of aristolochic acid IVa (AAIVa), which exists at higher levels in many Aristolochiaceae herbs, is extremely similar to AAI. In lack of toxicological data, it is unknown whether AAIVa exposure leads to aristolochic acid nephropathy (AAN), mutations, and tumorigenesis as of AAI. To answer these questions, mice were administered AAIVa by single or repeated long-term gavage, while AAI was used as a positive control. We found that single gavage of 40 mg/kg of AAIVa exhibited no obvious toxicity. Also, there were no tumors or death in mice administrated with 1 and 10 mg/kg of AAIVa for 6 months followed by a 12-month recovery time. There were no noteworthy alterations in gene mutation frequency in the kidney, liver, and stomach between the AAIVa and control mice. Fascinatingly, AA-associated mutational signatures, adenine-to-thymine (A>T) transversions, were absent in AAIVa-treated mice. Nonetheless, 10 mg/kg of AAIVa triggered lymphocytic infiltration and slight fibrous hyperplasia in the kidney at the 6th month; however, these were alleviated at the 12th and 18th months. On the contrary, AAI (positive control) caused severe diffuse fibrosis, tubular atrophy, necrosis, tumors in the forestomach and kidney, and death after the 6th month. It seems that long-term AAIVa exposure induced mild renal lesions could be due to the activation of the canonical or noncanonical transforming growth factor-ß (TGFß) pathway. Overall, these findings suggest that the mutagenicity and carcinogenic risk of AAIVa are very low.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Enfermedades Renales/inducido químicamente , Animales , Ácidos Aristolóquicos/administración & dosificación , Ácidos Aristolóquicos/química , Carcinógenos/administración & dosificación , Carcinógenos/química , Carcinógenos/toxicidad , Relación Dosis-Respuesta a Droga , Femenino , Enfermedades Renales/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutágenos/administración & dosificación , Mutágenos/química , Mutágenos/toxicidad , Factores de Tiempo
9.
Arch Toxicol ; 95(8): 2839-2850, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34223934

RESUMEN

Aristolochic acids (AAs) are a family of natural compounds with AA I and AA II being known carcinogens, whose bioactivation causes DNA adducts formation. However, other congeners have rarely been investigated. This study aimed to investigate genotoxicity of AA IVa, which differs from AA I by a hydroxyl group, abundant in Aristolochiaceae plants. AA IVa reacted with 2'-deoxyadenosine (dA) and 2'-deoxyguanosine (dG) to form three dA and five dG adducts as identified by high-resolution mass spectrometry, among which two dA and three dG adducts were detected in reactions of AA IVa with calf thymus DNA (CT DNA). However, no DNA adducts were detected in the kidney, liver, and forestomach of orally dosed mice at 40 mg/kg/day for 2 days, and bone marrow micronucleus assay also yielded negative results. Pharmacokinetic analyses of metabolites in plasma indicated that AA IVa was mainly O-demethylated to produce a metabolite with two hydroxyl groups, probably facilitating its excretion. Meanwhile, no reduced metabolites were detected. The competitive reaction of AA I and AA IVa with CT DNA, with adducts levels varying with pH of reaction revealed that AA IVa was significantly less reactive than AA I, probably by hydroxyl deprotonation of AA IVa, which was explained by theoretical calculations for reaction barriers, energy levels of the molecular orbits, and charges at the reaction sites. In brief, although it could form DNA adducts in vitro, AA IVa was non-genotoxic in vivo, which was attributed to its low reactivity and biotransformation into an easily excreted metabolite rather than bioactivation.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Aductos de ADN/efectos de los fármacos , Daño del ADN/efectos de los fármacos , ADN/efectos de los fármacos , Animales , Ácidos Aristolóquicos/administración & dosificación , Ácidos Aristolóquicos/química , Carcinógenos/administración & dosificación , Carcinógenos/química , Carcinógenos/toxicidad , Concentración de Iones de Hidrógeno , Masculino , Ratones , Ratones Endogámicos C57BL , Pruebas de Micronúcleos , Pruebas de Mutagenicidad
10.
Anticancer Res ; 41(8): 3833-3842, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34281843

RESUMEN

BACKGROUND/AIM: Lung cancer is the leading cause of cancer death worldwide. Cigarette smoke is the most important risk factor for cancer development. Growing evidence indicates that prolonged nicotine exposure is a potential factor associated with tumorigenesis. Here, the effect of prolonged nicotine exposure on A549 cells was investigated, using label-free quantitative proteomics. MATERIALS AND METHODS: Selection of an invasive subpopulation from the A549 cell line was performed to reveal the differential expression of proteins in relation to prolonged nicotine exposure, using Boyden chamber assays in combination with a proteomics approach. RESULTS: One hundred proteins from the NicoA549-L5 subline showed significant change in expression compared to those from the A549-L5 subline and their A549 parental cell line. Heat shock protein, protein disulfide isomerase A3, profilin-1 and legumain were expressed at higher levels in A549 cells after prolonged nicotine exposure. CONCLUSION: These aberrant proteins might serve as novel cancer biomarkers for cigarette smokers.


Asunto(s)
Nicotina/toxicidad , Proteínas/metabolismo , Proteómica/métodos , Células A549 , Biomarcadores de Tumor/metabolismo , Carcinógenos/administración & dosificación , Carcinógenos/toxicidad , Cisteína Endopeptidasas/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Nicotina/administración & dosificación , Profilinas/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem/métodos
11.
Arch Toxicol ; 95(8): 2851-2865, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34160648

RESUMEN

Chromosome aberrations (CAs), i.e. changes in chromosome number or structure, are known to cause chromosome rearrangements and subsequently tumorigenesis. However, the involvement of CAs in chemical-induced carcinogenesis is unclear. In the current study, we aimed to clarify the possible involvement of CAs in chemical carcinogenesis using a rat model with the non-mutagenic hepatocarcinogen acetamide. In an in vivo micronucleus (MN) test, acetamide was revealed to induce CAs specifically in rat liver at carcinogenic doses. Acetamide also induced centromere-positive large MN (LMN) in hepatocytes. Immunohistochemical and electron microscopic analyses of the LMN, which can be histopathologically detected as basophilic cytoplasmic inclusion, revealed abnormal expression of nuclear envelope proteins, increased heterochromatinization, and massive DNA damage. These molecular pathological features in LMN progressed with acetamide exposure in a time-dependent manner, implying that LMN formation can lead to chromosome rearrangements. Overall, these data suggested that CAs induced by acetamide play a pivotal role in acetamide-induced hepatocarcinogenesis in rats and that CAs can cause chemical carcinogenesis in animals via MN formation.


Asunto(s)
Acetamidas/toxicidad , Aberraciones Cromosómicas/inducido químicamente , Hepatocitos/efectos de los fármacos , Neoplasias Hepáticas/inducido químicamente , Acetamidas/administración & dosificación , Animales , Carcinogénesis/inducido químicamente , Carcinógenos/administración & dosificación , Carcinógenos/toxicidad , Hepatocitos/patología , Neoplasias Hepáticas/patología , Masculino , Pruebas de Micronúcleos , Ratas , Ratas Endogámicas F344 , Factores de Tiempo
12.
Arch Toxicol ; 95(8): 2883-2889, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34148101

RESUMEN

The International Agency for Research on Cancer (IARC) has recently proposed employing "ten key characteristics of human carcinogens" (TKCs) to determine the potential of agents for harmful effects. The TKCs seem likely to confuse the unsatisfactory correlation from testing regimes that have ignored the differences evident when cellular changes are compared in short and long-lived species, with their very different stem cell and somatic cell phylogenies. The proposed characteristics are so broad that their use will lead to an increase in the current unacceptably high rate of false positives. It could be an informative experiment to take well-established approved therapeutics with well-known human safety profiles and test them against this new TKC paradigm. Cancers are initiated and driven by heritable and transient changes in gene expression, expand clonally, and progress via additional associated acquired mutations and epigenetic alterations that provide cells with an evolutionary advantage. The genotoxicity testing protocols currently employed and required by regulation, emphasize testing for the mutational potential of the test agent. Two-year, chronic rodent cancer bioassays are intended to test for the entire spectrum of carcinogenic transformation. The use of cytotoxic doses causing increased, sustained cell proliferation that facilitates accumulated genetic damage leads to a high false-positive rate of tumor induction. Current cancer hazard assessment protocols and weight-of-the-evidence analysis of agent-specific cancer risk align poorly with the pathogenesis of human carcinoma and so need modernization and improvement in ways suggested here.


Asunto(s)
Carcinogénesis/inducido químicamente , Carcinógenos/toxicidad , Neoplasias/inducido químicamente , Animales , Pruebas de Carcinogenicidad/métodos , Carcinógenos/administración & dosificación , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Pruebas de Mutagenicidad/métodos , Medición de Riesgo , Roedores , Sensibilidad y Especificidad
13.
Regul Toxicol Pharmacol ; 124: 104977, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34174380

RESUMEN

The National Toxicology Program (NTP) reported that chronic dietary exposure to 4-methylimidazole (4-MeI) increased the incidence of lung adenomas/carcinomas beyond the normally high spontaneous rate in B6C3F1 mice. To examine plausible modes of action (MoAs) for mouse lung tumors (MLTs) upon exposure to high levels of 4-MeI, and their relevance in assessing human risk, a systematic approach was used to identify and evaluate mechanistic data (in vitro and in vivo) in the primary and secondary literature, along with high-throughput screening assay data. Study quality, relevance, and activity of mechanistic data identified across the evidence-base were organized according to key characteristics of carcinogens (KCCs) to identify potential key events in known or novel MLT MoAs. Integration of these evidence streams provided confirmation that 4-MeI lacks genotoxic and cytotoxic activity with some evidence to support a lack of mitogenic activity. Further evaluation of contextual and chemical-specific characteristics of 4-MeI was consequently undertaken. Due to lack of genotoxicity, along with transcriptomic and histopathological lung changes up to 28 and 90 days of exposure, the collective evidence suggests MLTs observed following exposure to high levels of 4-MeI develop at a late stage in the mouse chronic bioassay, albeit the exact MoA remains unclear.


Asunto(s)
Carcinógenos/toxicidad , Imidazoles/toxicidad , Neoplasias Pulmonares/epidemiología , Neoplasias Experimentales/epidemiología , Pruebas de Toxicidad Crónica/estadística & datos numéricos , Animales , Carcinógenos/administración & dosificación , Interpretación Estadística de Datos , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Imidazoles/administración & dosificación , Incidencia , Pulmón/efectos de los fármacos , Pulmón/patología , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/patología , Ratones , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/patología , Medición de Riesgo/métodos , Medición de Riesgo/estadística & datos numéricos , Pruebas de Toxicidad Crónica/métodos
14.
Toxicology ; 457: 152806, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33961948

RESUMEN

Colorectal cancer (CRC) is the third most common cancer worldwide and the second leading cause of cancer death. Benzo[a]pyrene (BaP) and 2-amino-1-methyl-6-phenylimidazol [4,5-b] pyridine (PhIP) present in cooked meat are pro-carcinogens and considered to be potential risk factors for CRC. Their carcinogenic and mutagenic effects require metabolic activation primarily by cytochrome P450 1 family enzymes (CYPs); the expression of these enzymes can be modulated by aryl hydrocarbon receptor (AhR) activation and the tumour microenvironment, involving mediators of inflammation. In this study, we hypothesized that tumour necrosis factor-α (TNF-α), a key mediator of inflammation, modulates BaP- and PhIP-induced DNA damage in colon cancer epithelial cells. Importantly, we observed that TNF-α alone (0.1-100 pg/ml) induced DNA damage (micronuclei formation) in HCT-116 cells and co-treatment of TNF-α with BaP or PhIP showed higher levels of DNA damage compared to the individual single treatments. TNF-α alone or in combination with BaP or PhIP did not affect the expression levels of CYP1A1 and CYP1B1 (target genes of AhR signaling pathways). The DNA damage induced by TNF-α was elevated in p53 null HTC-116 cells compared to wild type cells, suggesting that TNF-α-induced DNA damage is suppressed by functional p53. In contrast, p53 status failed to affect BaP and PhIP induced micronucleus frequency. Furthermore, JNK and NF-κB signaling pathway were activated by TNF-α treatment but only inhibition of JNK significantly reduced TNF-α-induced DNA damage. Collectively, these findings suggest that TNF-α induced DNA damage involves JNK signaling pathway rather than AhR and NF-κB pathways in colon cancer epithelial cells.


Asunto(s)
Carcinógenos/toxicidad , Neoplasias Colorrectales/metabolismo , Daño del ADN/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factor de Necrosis Tumoral alfa/toxicidad , Carcinógenos/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Neoplasias Colorrectales/patología , Daño del ADN/fisiología , Relación Dosis-Respuesta a Droga , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células HCT116 , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Factor de Necrosis Tumoral alfa/administración & dosificación
15.
Arch Toxicol ; 95(6): 2189-2199, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33938965

RESUMEN

Aristolochic acid (AA-I) induces upper urothelial tract cancer (UUTC) and bladder cancer (BC) in humans. AA-I forms the 7-(2'-deoxyadenosin-N6-yl)aristolactam I (dA-AL-I) adduct, which induces multiple A:T-to-T:A transversion mutations in TP53 of AA-I exposed UTUC patients. This mutation is rarely reported in TP53 of other transitional cell carcinomas and thus recognized as an AA-I mutational signature. A:T-to-T:A transversion mutations were recently detected in bladder tumors of patients in Asia with known AA-I-exposure, implying that AA-I contributes to BC. Mechanistic studies on AA-I genotoxicity have not been reported in human bladder. In this study, we examined AA-I DNA adduct formation and mechanisms of toxicity in the human RT4 bladder cell line. The biological potencies of AA-I were compared to 4-aminobiphenyl, a recognized human bladder carcinogen, and several structurally related carcinogenic heterocyclic aromatic amines (HAA), which are present in urine of smokers and omnivores. AA-I (0.05-10 µM) induced a concentration- and time-dependent cytotoxicity. AA-I (100 nM) DNA adduct formation occurred at over a thousand higher levels than the principal DNA adducts formed with 4-ABP or HAAs (1 µM). dA-AL-I adduct formation was detected down to a 1 nM concentration. Studies with selective chemical inhibitors provided evidence that NQO1 is the major enzyme involved in AA-I bio-activation in RT4 cells, whereas CYP1A1, another enzyme implicated in AA-I toxicity, had a lesser role in bio-activation or detoxification of AA-I. AA-I DNA damage also induced genotoxic stress leading to p53-dependent apoptosis. These biochemical data support the human mutation data and a role for AA-I in BC.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Carcinógenos/toxicidad , Daño del ADN/efectos de los fármacos , Vejiga Urinaria/efectos de los fármacos , Compuestos de Aminobifenilo/toxicidad , Ácidos Aristolóquicos/administración & dosificación , Carcinógenos/administración & dosificación , Línea Celular Tumoral , Citocromo P-450 CYP1A1/metabolismo , Aductos de ADN/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Mutación , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Proteína p53 Supresora de Tumor/genética , Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/patología
16.
J Mammary Gland Biol Neoplasia ; 26(2): 113-120, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33866475

RESUMEN

RNAscope is a quantitative in situ gene expression measurement technique that preserves the spatial aspect of intact tissue; thus, allowing for comparison of specific cell populations and morphologies. Reliable and accurate measurement of gene expression in tissue is dependent on preserving RNA integrity and the quantitative nature of RNAscope. The purpose of this study was to determine if the quantitative nature of RNAscope was retained following processing and carmine staining of mammary gland whole-mounts, which are commonly used to identify lesions, such as hyperplasia and ductal carcinoma in situ (DCIS). We were concerned that handling and procedures required to visualize microscopic disease lesions might compromise RNA integrity and the robustness of RNAscope. No effect on the quantitative abilities of RNAscope was detected when mammary gland whole-mounts were pre-screened for lesions of interest prior to RNAscope. This was determined in comparison to tissue that had been formalin-fixed and paraffin embedded (FFPE) immediately after collection. The ability to pre-screen whole-mounts allowed unpalpable diseased lesions to be identified without labor-intensive serial sectioning of tissue samples to find diseased tissue. This method is applicable to evaluate mammary gland whole-mounts during normal mammary gland development, function, and disease progression.


Asunto(s)
Carcinoma Intraductal no Infiltrante/diagnóstico , Perfilación de la Expresión Génica/métodos , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/diagnóstico , 9,10-Dimetil-1,2-benzantraceno/administración & dosificación , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Carcinógenos/administración & dosificación , Carcinógenos/toxicidad , Carcinoma Intraductal no Infiltrante/inducido químicamente , Carcinoma Intraductal no Infiltrante/genética , Carcinoma Intraductal no Infiltrante/patología , Modelos Animales de Enfermedad , Femenino , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , ARN/metabolismo , Ratas , Conservación de Tejido/métodos
17.
Drug Res (Stuttg) ; 71(6): 341-347, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33862663

RESUMEN

Farnesyltransferase (FTase) is a zinc enzyme that has been the subject of attention in anti-cancer research over the past. In this study, phytochemicals from Curcuma longa L., Taraxacum officinale, and Spondias mombin plants were screened for their inhibitory potentials on the human farnesyltransferase. A three-dimensional quantitative structure-activity relationship (3D-QSAR) model for the inhibition of farnesyltransferase was generated and the inhibition of farnesyltransferase by the hit, ascorbic acid was validated in an animal model of breast cancer. The lead compound, ascorbic acid makes extensive hydrogen bond interactions with key residues, lys-353, tyr-300, gly-290, leu-290 within the active site of farnesyltransferase. It downregulated the expression of FNTA mRNA in an animal model of breast cancer. The 3D-QSAR generated herein is robust, thoroughly validated, and should be employed in the pipelining of novel farnesyltransferase inhibitors. Ascorbic acid demonstrates its anticancer potentials through the inhibition of farnesyltransferase.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Ácido Ascórbico/farmacología , Inhibidores Enzimáticos/farmacología , Farnesiltransferasa/antagonistas & inhibidores , Neoplasias Mamarias Experimentales/tratamiento farmacológico , 9,10-Dimetil-1,2-benzantraceno/administración & dosificación , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Anacardiaceae/química , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/uso terapéutico , Ácido Ascórbico/química , Ácido Ascórbico/uso terapéutico , Carcinógenos/administración & dosificación , Carcinógenos/toxicidad , Curcuma/química , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/uso terapéutico , Farnesiltransferasa/metabolismo , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Concentración 50 Inhibidora , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/patología , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad Cuantitativa , Ratas , Taraxacum/química
18.
Chem Biol Interact ; 341: 109464, 2021 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-33823170

RESUMEN

Current regulatory cancer risk assessment principles and practices assume a linear dose-response relationship-the linear no-threshold (LNT) model-that theoretically estimates cancer risks occurring following low doses of carcinogens by linearly extrapolating downward from experimentally determined risks at high doses. The two-year rodent bioassays serve as experimental vehicles to determine the high-dose cancer risks in animals and then to predict, by extrapolation, the number of carcinogen-induced tumors (tumor incidence) that will arise during the lifespans of humans who are exposed to environmental carcinogens at doses typically orders of magnitude below those applied in the rodent assays. An integrated toxicological analysis is conducted herein to reconsider an alternative and once-promising approach, tumor latency, for estimating carcinogen-induced cancer risks at low doses. Tumor latency measures time-to-tumor following exposure to a carcinogen, instead of tumor incidence. Evidence for and against the concept of carcinogen-induced tumor latency is presented, discussed, and then examined with respect to its relationship to dose, dose rates, and the dose-related concepts of initiation, tumor promotion, tumor regression, tumor incidence, and hormesis. Considerable experimental evidence indicates: (1) tumor latency (time-to-tumor) is inversely related to the dose of carcinogens and (2) lower doses of carcinogens display quantifiably discrete latency thresholds below which the promotion and, consequently, the progression and growth of tumors are delayed or prevented during a normal lifespan. Besides reconciling well with the concept of tumor promotion, such latency thresholds also reconcile favorably with the existence of thresholds for tumor incidence, the stochastic processes of tumor initiation, and the compensatory repair mechanisms of hormesis. Most importantly, this analysis and the arguments presented herein provide sound theoretical, experimental, and mechanistic rationales for rethinking the foundational premises of low-dose linearity and updating the current practices of cancer risk assessment to include the concept of carcinogen thresholds.


Asunto(s)
Pruebas de Carcinogenicidad/métodos , Carcinógenos/administración & dosificación , Carcinógenos/toxicidad , Neoplasias/inducido químicamente , Animales , Relación Dosis-Respuesta a Droga , Hormesis , Humanos , Incidencia , Neoplasias/epidemiología , Medición de Riesgo/métodos , Pruebas de Toxicidad Crónica/métodos
19.
Chem Res Toxicol ; 34(3): 723-732, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33629582

RESUMEN

Tobacco smoke is a complex mixture of chemicals, many of which are toxic and carcinogenic. Hazard assessments of tobacco smoke exposure have predominantly focused on either single chemical exposures or the more complex mixtures of tobacco smoke or its fractions. There are fewer studies exploring interactions between specific tobacco smoke chemicals. Aldehydes such as formaldehyde and acetaldehyde were hypothesized to enhance the carcinogenic properties of the human carcinogen, 4-methylnitrosamino-1-(3-pyridyl)-1-butanone (NNK) through a variety of mechanisms. This hypothesis was tested in the established NNK-induced A/J mouse lung tumor model. A/J mice were exposed to NNK (intraperitoneal injection, 0, 2.5, or 7.5 µmol in saline) in the presence or absence of acetaldehyde (0 or 360 ppmv) or formaldehyde (0 or 17 ppmv) for 3 h in a nose-only inhalation chamber, and lung tumors were counted 16 weeks later. Neither aldehyde by itself induced lung tumors. However, mice receiving both NNK and acetaldehyde or formaldehyde had more adenomas with dysplasia or progression than those receiving only NNK, suggesting that aldehydes may increase the severity of NNK-induced lung adenomas. The aldehyde coexposure did not affect the levels of NNK-derived DNA adduct levels. Similar studies tested the ability of a 3 h nose-only carbon dioxide (0, 5, 10, or 15%) coexposure to influence lung adenoma formation by NNK. While carbon dioxide alone was not carcinogenic, it significantly increased the number of NNK-derived lung adenomas without affecting NNK-derived DNA damage. These studies indicate that the chemicals in tobacco smoke work together to form a potent lung carcinogenic mixture.


Asunto(s)
Aldehídos/toxicidad , Dióxido de Carbono/toxicidad , Carcinógenos/toxicidad , Neoplasias Pulmonares/inducido químicamente , Nitrosaminas/toxicidad , Administración por Inhalación , Aldehídos/administración & dosificación , Aldehídos/química , Animales , Dióxido de Carbono/administración & dosificación , Dióxido de Carbono/química , Carcinógenos/administración & dosificación , Carcinógenos/química , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Neoplasias Pulmonares/metabolismo , Ratones , Estructura Molecular , Nitrosaminas/administración & dosificación , Nicotiana/química
20.
Regul Toxicol Pharmacol ; 120: 104858, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33387565

RESUMEN

Dichloromethane (DCM) is a high production volume chemical (>1000 t/a) mainly used as an industrial solvent. Carcinogenicity studies in rats, mice and hamsters have demonstrated a malignant tumor inducing potential of DCM only in the mouse (lung and liver) at 1000-4000 ppm whereas human data do not support a conclusion of cancer risk. Based on this, DCM has been classified as a cat. 2 carcinogen. Dose-dependent toxicokinetics of DCM suggest that DCM is a threshold carcinogen in mice, initiating carcinogenicity via the low affinity/high capacity GSTT1 pathway; a biotransformation pathway that becomes relevant only at high exposure concentrations. Rats and hamsters have very low activities of this DCM-metabolizing GST and humans have even lower activities of this enzyme. Based on the induction of specific tumors selectively in the mouse, the dose- and species-specific toxicokinetics in this species, and the absence of a malignant tumor response by DCM in rats and hamsters having a closer relationship to DCM toxicokinetics in humans and thus being a more relevant animal model, the current classification of DCM as human carcinogen cat. 2 remains appropriate.


Asunto(s)
Carcinógenos/administración & dosificación , Carcinógenos/toxicidad , Modelos Animales de Enfermedad , Cloruro de Metileno/administración & dosificación , Cloruro de Metileno/toxicidad , Administración por Inhalación , Animales , Biotransformación/efectos de los fármacos , Biotransformación/fisiología , Cricetinae , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/normas , Humanos , Ratones , Ratas , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA