Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
EFSA J ; 19(8): e06768, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34377190

RESUMO

The EFSA has updated the Guidance on risk assessment of the application of nanoscience and nanotechnologies in the food and feed chain, human and animal health. It covers the application areas within EFSA's remit, including novel foods, food contact materials, food/feed additives and pesticides. The updated guidance, now Scientific Committee Guidance on nano risk assessment (SC Guidance on Nano-RA), has taken account of relevant scientific studies that provide insights to physico-chemical properties, exposure assessment and hazard characterisation of nanomaterials and areas of applicability. Together with the accompanying Guidance on Technical requirements for regulated food and feed product applications to establish the presence of small particles including nanoparticles (Guidance on Particle-TR), the SC Guidance on Nano-RA specifically elaborates on physico-chemical characterisation, key parameters that should be measured, methods and techniques that can be used for characterisation of nanomaterials and their determination in complex matrices. The SC Guidance on Nano-RA also details aspects relating to exposure assessment and hazard identification and characterisation. In particular, nanospecific considerations relating to in vitro/in vivo toxicological studies are discussed and a tiered framework for toxicological testing is outlined. Furthermore, in vitro degradation, toxicokinetics, genotoxicity, local and systemic toxicity as well as general issues relating to testing of nanomaterials are described. Depending on the initial tier results, additional studies may be needed to investigate reproductive and developmental toxicity, chronic toxicity and carcinogenicity, immunotoxicity and allergenicity, neurotoxicity, effects on gut microbiome and endocrine activity. The possible use of read-across to fill data gaps as well as the potential use of integrated testing strategies and the knowledge of modes or mechanisms of action are also discussed. The Guidance proposes approaches to risk characterisation and uncertainty analysis.

2.
Nucleic Acids Res ; 48(4): 1652-1668, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-31930303

RESUMO

The excision of mutagenic DNA adducts by the nucleotide excision repair (NER) pathway is essential for genome stability, which is key to avoiding genetic diseases, premature aging, cancer and neurologic disorders. Due to the need to process an extraordinarily high damage density embedded in the nucleosome landscape of chromatin, NER activity provides a unique functional caliper to understand how histone modifiers modulate DNA damage responses. At least three distinct lysine methyltransferases (KMTs) targeting histones have been shown to facilitate the detection of ultraviolet (UV) light-induced DNA lesions in the difficult to access DNA wrapped around histones in nucleosomes. By methylating core histones, these KMTs generate docking sites for DNA damage recognition factors before the chromatin structure is ultimately relaxed and the offending lesions are effectively excised. In view of their function in priming nucleosomes for DNA repair, mutations of genes coding for these KMTs are expected to cause the accumulation of DNA damage promoting cancer and other chronic diseases. Research on the question of how KMTs modulate DNA repair might pave the way to the development of pharmacologic agents for novel therapeutic strategies.


Assuntos
Cromatina/genética , Dano ao DNA/genética , Histona Metiltransferases/genética , Histonas/genética , Cromatina/efeitos da radiação , Dano ao DNA/efeitos da radiação , Reparo do DNA/genética , Reparo do DNA/efeitos da radiação , Instabilidade Genômica/genética , Instabilidade Genômica/efeitos da radiação , Histona Metiltransferases/química , Metilação/efeitos da radiação , Nucleossomos/genética , Nucleossomos/efeitos da radiação , Saccharomyces cerevisiae/genética , Transdução de Sinais/efeitos da radiação , Raios Ultravioleta
3.
BMC Cancer ; 19(1): 1042, 2019 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-31690264

RESUMO

BACKGROUND: DNA-crosslinking agents like cisplatin and mitomycin C (MMC) are indispensible for the treatment of many solid malignancies. These anticancer drugs generate DNA interstrand crosslinks (ICLs) that cause cell death by blocking replication forks. Many factors counteracting ICL-induced DNA replication stress, including the Fanconi anemia (FA) pathway, are regulated by ubiquitination and, therefore, ubiquitin ligases are potential targets for the sensitization of cancer cells to crosslinking agents. In this study, we investigated the function of the CRL4 ubiquitin ligase in modulating the response of cancer cells to ICL induction. METHODS: The two cullin paralogs CUL4A and CUL4B, which form the CRL4 ligase scaffold, were depleted in cancer cells by small interfering RNA followed by analysis of the cellular and biochemical responses to ICLs elicited upon cisplatin or MMC treatment. RESULTS: We report that the combined depletion of CUL4A and CUL4B weakens an FA pathway-dependent S phase checkpoint response. CRL4 positively stimulates the monoubiquitination of FANCD2 required for the recruitment of XPF-ERCC1, a structure-specific endonuclease that, in turn, contributes to the display of single-stranded DNA (ssDNA) at ICLs. After CRL4 down regulation, the missing ssDNA results in reduced recruitment of RPA, thereby dampening activation of ATR and CHK1 checkpoint kinases and allowing for S phase progression despite ICL induction. CONCLUSION: Our findings indicate that CRL4 promotes cell survival by potentiating an FA pathway-dependent ssDNA-RPA signaling platform installed at ICLs. The anticancer efficacy of crosslinking agents may, therefore, be enhanced by down regulating CRL4 activity.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , DNA de Cadeia Simples/genética , Anemia de Fanconi/metabolismo , Mitomicina/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Proteínas Culina/genética , Dano ao DNA , Feminino , Humanos , Neoplasias Ovarianas/genética , RNA Interferente Pequeno/genética , Pontos de Checagem da Fase S do Ciclo Celular , Transdução de Sinais , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
4.
EFSA J ; 17(6): e05708, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32626331

RESUMO

The Scientific Committee confirms that the Threshold of Toxicological Concern (TTC) is a pragmatic screening and prioritisation tool for use in food safety assessment. This Guidance provides clear step-by-step instructions for use of the TTC approach. The inclusion and exclusion criteria are defined and the use of the TTC decision tree is explained. The approach can be used when the chemical structure of the substance is known, there are limited chemical-specific toxicity data and the exposure can be estimated. The TTC approach should not be used for substances for which EU food/feed legislation requires the submission of toxicity data or when sufficient data are available for a risk assessment or if the substance under consideration falls into one of the exclusion categories. For substances that have the potential to be DNA-reactive mutagens and/or carcinogens based on the weight of evidence, the relevant TTC value is 0.0025 µg/kg body weight (bw) per day. For organophosphates or carbamates, the relevant TTC value is 0.3 µg/kg bw per day. All other substances are grouped according to the Cramer classification. The TTC values for Cramer Classes I, II and III are 30 µg/kg bw per day, 9 µg/kg bw per day and 1.5 µg/kg bw per day, respectively. For substances with exposures below the TTC values, the probability that they would cause adverse health effects is low. If the estimated exposure to a substance is higher than the relevant TTC value, a non-TTC approach is required to reach a conclusion on potential adverse health effects.

5.
J Nanobiotechnology ; 16(1): 51, 2018 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-29859103

RESUMO

Nanomaterial engineering provides an important technological advance that offers substantial benefits for applications not only in the production and processing, but also in the packaging and storage of food. An expanding commercialization of nanomaterials as part of the modern diet will substantially increase their oral intake worldwide. While the risk of particle inhalation received much attention, gaps of knowledge exist regarding possible adverse health effects due to gastrointestinal exposure. This problem is highlighted by pigment-grade titanium dioxide (TiO2), which confers a white color and increased opacity with an optimal particle diameter of 200-300 nm. However, size distribution analyses showed that batches of food-grade TiO2 always comprise a nano-sized fraction as inevitable byproduct of the manufacturing processes. Submicron-sized TiO2 particles, in Europe listed as E 171, are widely used as a food additive although the relevant risk assessment has never been satisfactorily completed. For example, it is not possible to derive a safe daily intake of TiO2 from the available long-term feeding studies in rodents. Also, the use of TiO2 particles in the food sector leads to highest exposures in children, but only few studies address the vulnerability of this particular age group. Extrapolation of animal studies to humans is also problematic due to knowledge gaps as to local gastrointestinal effects of TiO2 particles, primarily on the mucosa and the gut-associated lymphoid system. Tissue distributions after oral administration of TiO2 differ from other exposure routes, thus limiting the relevance of data obtained from inhalation or parenteral injections. Such difficulties and uncertainties emerging in the retrospective assessment of TiO2 particles exemplify the need for a fit-to-purpose data requirement for the future evaluation of novel nano-sized or submicron-sized particles added deliberately to food.


Assuntos
Aditivos Alimentares/toxicidade , Titânio/toxicidade , Animais , Células Cultivadas , Exposição Ambiental , Humanos , Camundongos , Testes de Toxicidade
6.
EFSA J ; 16(3): e05082, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32625822

RESUMO

Moniliformin (MON) is a mycotoxin with low molecular weight primarily produced by Fusarium fungi and occurring predominantly in cereal grains. Following a request of the European Commission, the CONTAM Panel assessed the risk of MON to human and animal health related to its presence in food and feed. The limited information available on toxicity and on toxicokinetics in experimental and farm animals indicated haematotoxicity and cardiotoxicity as major adverse health effects of MON. MON causes chromosome aberrations in vitro but no in vivo genotoxicity data and no carcinogenicity data were identified. Due to the limitations in the available toxicity data, human acute or chronic health-based guidance values (HBGV) could not be established. The margin of exposure (MOE) between the no-observed-adverse-effect level (NOAEL) of 6.0 mg/kg body weight (bw) for cardiotoxicity from a subacute study in rats and the acute upper bound (UB) dietary exposure estimates ranged between 4,000 and 73,000. The MOE between the lowest benchmark dose lower confidence limit (for a 5% response - BMDL05) of 0.20 mg MON/kg bw per day for haematological hazards from a 28-day study in pigs and the chronic dietary human exposure estimates ranged between 370 and 5,000,000 for chronic dietary exposures. These MOEs indicate a low risk for human health but were associated with high uncertainty. The toxicity data available for poultry, pigs, and mink indicated a low or even negligible risk for these animals from exposure to MON in feed at the estimated exposure levels under current feeding practices. Assuming similar or lower sensitivity as for pigs, the CONTAM Panel considered a low or even negligible risk for the other animal species for which no toxicity data suitable for hazard characterisation were identified. Additional toxicity studies are needed and depending on their outcome, the collection of more occurrence data on MON in food and feed is recommended to enable a comprehensive human risk assessment.

7.
Nat Commun ; 8(1): 1333, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-29109511

RESUMO

Global-genome nucleotide excision repair (GG-NER) prevents ultraviolet (UV) light-induced skin cancer by removing mutagenic cyclobutane pyrimidine dimers (CPDs). These lesions are formed abundantly on DNA wrapped around histone octamers in nucleosomes, but a specialized damage sensor known as DDB2 ensures that they are accessed by the XPC initiator of GG-NER activity. We report that DDB2 promotes CPD excision by recruiting the histone methyltransferase ASH1L, which methylates lysine 4 of histone H3. In turn, methylated H3 facilitates the docking of the XPC complex to nucleosomal histone octamers. Consequently, DDB2, ASH1L and XPC proteins co-localize transiently on histone H3-methylated nucleosomes of UV-exposed cells. In the absence of ASH1L, the chromatin binding of XPC is impaired and its ability to recruit downstream GG-NER effectors diminished. Also, ASH1L depletion suppresses CPD excision and confers UV hypersensitivity. These findings show that ASH1L configures chromatin for the effective handoff between damage recognition factors during GG-NER activity.


Assuntos
Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Células HeLa , Histona-Lisina N-Metiltransferase , Histonas/metabolismo , Humanos , Metilação , Nucleossomos/metabolismo , Domínios e Motivos de Interação entre Proteínas , Dímeros de Pirimidina/metabolismo , RNA Interferente Pequeno/genética , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/prevenção & controle , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Raios Ultravioleta/efeitos adversos
8.
EMBO J ; 36(22): 3372-3386, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29018037

RESUMO

Ultraviolet (UV) light induces mutagenic cyclobutane pyrimidine dimers (CPDs) in nucleosomal DNA that is tightly wrapped around histone octamers. How global-genome nucleotide excision repair (GG-NER) processes CPDs despite that this chromatin arrangement is poorly understood. An increased chromatin association of CHD1 (chromodomain helicase DNA-binding 1) upon UV irradiation indicated possible roles of this chromatin remodeler in the UV damage response. Immunoprecipitation of chromatin fragments revealed that CHD1 co-localizes in part with GG-NER factors. Chromatin fractionation showed that the UV-dependent recruitment of CHD1 occurs to UV lesions in histone-assembled nucleosomal DNA and that this CHD1 relocation requires the lesion sensor XPC (xeroderma pigmentosum group C). In situ immunofluorescence analyses further demonstrate that CHD1 facilitates substrate handover from XPC to the downstream TFIIH (transcription factor IIH). Consequently, CHD1 depletion slows down CPD excision and sensitizes cells to UV-induced cytotoxicity. The finding of a CHD1-driven lesion handover between sequentially acting GG-NER factors on nucleosomal histone octamers suggests that chromatin provides a recognition scaffold enabling the detection of a subset of CPDs.


Assuntos
Montagem e Desmontagem da Cromatina , Dano ao DNA , DNA Helicases/metabolismo , Reparo do DNA/efeitos da radiação , Proteínas de Ligação a DNA/metabolismo , Nucleossomos/metabolismo , Fator de Transcrição TFIIH/metabolismo , Raios Ultravioleta , Xeroderma Pigmentoso/metabolismo , Morte Celular/efeitos da radiação , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina/efeitos da radiação , Genoma Humano , Células HEK293 , Células HeLa , Humanos , Nucleossomos/efeitos da radiação , Dímeros de Pirimidina/metabolismo , RNA Interferente Pequeno/metabolismo
9.
Part Fibre Toxicol ; 14(1): 21, 2017 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-28645296

RESUMO

BACKGROUND: Dendritic cells (DCs) are specialized first-line sensors of foreign materials invading the organism. These sentinel cells rely on pattern recognition receptors such as Nod-like or Toll-like receptors (TLRs) to launch immune reactions against pathogens, but also to mediate tolerance to self-antigens and, in the intestinal milieu, to nutrients and commensals. Since inappropriate DC activation contributes to inflammatory diseases and immunopathologies, a key question in the evaluation of orally ingested nanomaterials is whether their contact with DCs in the intestinal mucosa disrupts this delicate homeostatic balance between pathogen defense and tolerance. Here, we generated steady-state DCs by incubating hematopoietic progenitors with feline McDonough sarcoma-like tyrosine kinase 3 ligand (Flt3L) and used the resulting immature DCs to test potential biological responses against food-grade synthetic amorphous silica (SAS) representing a common nanomaterial generally thought to be safe. RESULTS: Interaction of immature and unprimed DCs with food-grade SAS particles and their internalization by endocytic uptake fails to elicit cytotoxicity and the release of interleukin (IL)-1α or tumor necrosis factor-α, which were identified as master regulators of acute inflammation in lung-related studies. However, the display of maturation markers on the cell surface shows that SAS particles activate completely immature DCs. Also, the endocytic uptake of SAS particles into these steady-state DCs leads to induction of the pro-IL-1ß precursor, subsequently cleaved by the inflammasome to secrete mature IL-1ß. In contrast, neither pro-IL-1ß induction nor mature IL-1ß secretion occurs upon internalization of TiO2 or FePO4 nanoparticles. The pro-IL-1ß induction is suppressed by pharmacologic inhibitors of endosomal TLR activation or by genetic ablation of MyD88, a downstream adapter of TLR pathways, indicating that endosomal pattern recognition is responsible for the observed cytokine response to food-grade SAS particles. CONCLUSIONS: Our results unexpectedly show that food-grade SAS particles are able to directly initiate the endosomal MyD88-dependent pathogen pattern recognition and signaling pathway in steady-state DCs. The ensuing activation of immature DCs with de novo induction of pro-IL-1ß implies that the currently massive use of SAS particles as food additive should be reconsidered.


Assuntos
Células Dendríticas/efeitos dos fármacos , Aditivos Alimentares/toxicidade , Interleucina-1beta/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Precursores de Proteínas/metabolismo , Dióxido de Silício/toxicidade , Animais , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/ultraestrutura , Relação Dose-Resposta a Droga , Endocitose , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Endossomos/ultraestrutura , Aditivos Alimentares/síntese química , Aditivos Alimentares/metabolismo , Inocuidade dos Alimentos , Inflamassomos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Nanopartículas , Processamento de Proteína Pós-Traducional , Receptores de Reconhecimento de Padrão/metabolismo , Medição de Risco , Transdução de Sinais/efeitos dos fármacos , Dióxido de Silício/síntese química , Dióxido de Silício/metabolismo , Fatores de Tempo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Regulação para Cima
10.
Nanotoxicology ; 11(4): 496-506, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28368214

RESUMO

Nanotechnology offers new opportunities for providing health benefits in foods. Food fortification with iron phosphate nanoparticles (FePO4 NPs) is a promising new approach to reducing iron deficiency because FePO4 NPs combine high bioavailability with superior sensory performance in difficult to fortify foods. However, their safety remains largely untested. We fed rats for 90 days diets containing FePO4 NPs at doses at which iron sulfate (FeSO4), a commonly used food fortificant, has been shown to induce adverse effects. Feeding did not result in signs of toxicity, including oxidative stress, organ damage, excess iron accumulation in organs or histological changes. These safety data were corroborated by evidence that NPs were taken up by human gastrointestinal cell lines without reducing cell viability or inducing oxidative stress. Our findings suggest FePO4 NPs appear to be as safe for ingestion as FeSO4.


Assuntos
Compostos Férricos , Alimentos Fortificados , Nanopartículas , Estresse Oxidativo/efeitos dos fármacos , Animais , Disponibilidade Biológica , Sobrevivência Celular/efeitos dos fármacos , Dieta , Relação Dose-Resposta a Droga , Compostos Férricos/administração & dosagem , Compostos Férricos/efeitos adversos , Compostos Férricos/metabolismo , Glutationa/metabolismo , Células HT29 , Humanos , Sobrecarga de Ferro , Masculino , Nanopartículas/administração & dosagem , Nanopartículas/efeitos adversos , Nanopartículas/metabolismo , Ratos , Ratos Sprague-Dawley
11.
Chem Res Toxicol ; 30(3): 769-776, 2017 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-28076683

RESUMO

Bioactivation as well as DNA repair affects the susceptibility of cancer cells to the action of DNA-alkylating chemotherapeutic drugs. However, information is limited with regard to the relative contributions of these processes to the biological outcome of metabolically activated DNA alkylating agents. We evaluated the influence of cellular bioactivation capacity and DNA repair on cytotoxicity of the DNA alkylating agent acylfulvene (AF). We compared the cytotoxicity and RNA synthesis inhibition by AF and its synthetic activated analogue iso-M0 in a panel of fibroblast cell lines with deficiencies in transcription-coupled (TC-NER) or global genome nucleotide excision repair (GG-NER). We related these data to the inherent bioactivation capacity of each cell type on the basis of mRNA levels. We demonstrated that specific inactivation of TC-NER by siRNA had the largest positive impact on AF activity in a cancer cell line. These findings establish that transcription-coupled DNA repair reduces cellular sensitivity to AF, independent of the requirement for bioactivation.


Assuntos
Reparo do DNA , Sesquiterpenos/farmacologia , Compostos de Espiro/farmacologia , Transcrição Gênica/efeitos dos fármacos , Ativação Metabólica , Linhagem Celular , Humanos , Sesquiterpenos/farmacocinética , Compostos de Espiro/farmacocinética
12.
EFSA J ; 15(5): e04849, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-32625502

RESUMO

Following a request from the European Commission to EFSA, the EFSA Scientific Committee (SC) prepared a guidance for the risk assessment of substances present in food intended for infants below 16 weeks of age. In its approach to develop this guidance, the EFSA SC took into account, among others, (i) an exposure assessment based on infant formula as the only source of nutrition; (ii) knowledge of organ development in human infants, including the development of the gut, metabolic and excretory capacities, the brain and brain barriers, the immune system, the endocrine and reproductive systems; (iii) the overall toxicological profile of the substance identified through the standard toxicological tests, including critical effects; (iv) the relevance for the human infant of the neonatal experimental animal models used. The EFSA SC notes that during the period from birth up to 16 weeks, infants are expected to be exclusively fed on breast milk and/or infant formula. The EFSA SC views this period as the time where health-based guidance values for the general population do not apply without further considerations. High infant formula consumption per body weight is derived from 95th percentile consumption. The first weeks of life is the time of the highest relative consumption on a body weight basis. Therefore, when performing an exposure assessment, the EFSA SC proposes to use the high consumption value of 260 mL/kg bw per day. A decision tree approach is proposed that enables a risk assessment of substances present in food intended for infants below 16 weeks of age. The additional information needed when testing substances present in food for infants below 16 weeks of age and the approach to be taken for the risk assessment are on a case-by-case basis, depending on whether the substance is added intentionally to food and is systemically available.

13.
Front Genet ; 7: 68, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27200078

RESUMO

Global-genome nucleotide excision repair (GG-NER) prevents genome instability by excising a wide range of different DNA base adducts and crosslinks induced by chemical carcinogens, ultraviolet (UV) light or intracellular side products of metabolism. As a versatile damage sensor, xeroderma pigmentosum group C (XPC) protein initiates this generic defense reaction by locating the damage and recruiting the subunits of a large lesion demarcation complex that, in turn, triggers the excision of aberrant DNA by endonucleases. In the very special case of a DNA repair response to UV radiation, the function of this XPC initiator is tightly controlled by the dual action of cullin-type CRL4(DDB2) and sumo-targeted RNF111 ubiquitin ligases. This twofold protein ubiquitination system promotes GG-NER reactions by spatially and temporally regulating the interaction of XPC protein with damaged DNA across the nucleosome landscape of chromatin. In the absence of either CRL4(DDB2) or RNF111, the DNA excision repair of UV lesions is inefficient, indicating that these two ubiquitin ligases play a critical role in mitigating the adverse biological effects of UV light in the exposed skin.

14.
Cell Mol Life Sci ; 73(3): 547-66, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26521083

RESUMO

The cellular defense system known as global-genome nucleotide excision repair (GG-NER) safeguards genome stability by eliminating a plethora of structurally unrelated DNA adducts inflicted by chemical carcinogens, ultraviolet (UV) radiation or endogenous metabolic by-products. Xeroderma pigmentosum group C (XPC) protein provides the promiscuous damage sensor that initiates this versatile NER reaction through the sequential recruitment of DNA helicases and endonucleases, which in turn recognize and excise insulting base adducts. As a DNA damage sensor, XPC protein is very unique in that it (a) displays an extremely wide substrate range, (b) localizes DNA lesions by an entirely indirect readout strategy, (c) recruits not only NER factors but also multiple repair players, (d) interacts avidly with undamaged DNA, (e) also interrogates nucleosome-wrapped DNA irrespective of chromatin compaction and (f) additionally functions beyond repair as a co-activator of RNA polymerase II-mediated transcription. Many recent reports highlighted the complexity of a post-translational circuit that uses polypeptide modifiers to regulate the spatiotemporal activity of this multiuse sensor during the UV damage response in human skin. A newly emerging concept is that stringent regulation of the diverse XPC functions is needed to prioritize DNA repair while avoiding the futile processing of undamaged genes or silent genomic sequences.


Assuntos
Dano ao DNA , Reparo do DNA/genética , Proteínas de Ligação a DNA/fisiologia , Modelos Genéticos , Animais , Adutos de DNA , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Instabilidade Genômica , Humanos , Camundongos , Modelos Moleculares , Estrutura Terciária de Proteína , Neoplasias Cutâneas/genética , Raios Ultravioleta
15.
BMC Vet Res ; 10: 122, 2014 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-24888604

RESUMO

BACKGROUND: Hepatic lipidosis or fatty liver disease is a major metabolic disorder of high-producing dairy cows that compromises animal performance and, hence, causes heavy economic losses worldwide. This syndrome, occurring during the critical transition from gestation to early lactation, leads to an impaired health status, decreased milk yield, reduced fertility and shortened lifetime. Because the prevailing clinical chemistry parameters indicate advanced liver damage independently of the underlying disease, currently, hepatic lipidosis can only be ascertained by liver biopsy. We hypothesized that the condition of fatty liver disease may be accompanied by an altered profile of endogenous metabolites in the blood of affected animals. RESULTS: To identify potential small-molecule biomarkers as a novel diagnostic alternative, the serum samples of diseased dairy cows were subjected to a targeted metabolomics screen by triple quadrupole mass spectrometry. A subsequent multivariate test involving principal component and linear discriminant analyses yielded 29 metabolites (amino acids, phosphatidylcholines and sphingomyelines) that, in conjunction, were able to distinguish between dairy cows with no hepatic lipidosis and those displaying different stages of the disorder. CONCLUSIONS: This proof-of-concept study indicates that metabolomic profiles, including both amino acids and lipids, distinguish hepatic lipidosis from other peripartal disorders and, hence, provide a promising new tool for the diagnosis of hepatic lipidosis. By generating insights into the molecular pathogenesis of hepatic lipidosis, metabolomics studies may also facilitate the prevention of this syndrome.


Assuntos
Doenças dos Bovinos/sangue , Lipidoses/veterinária , Hepatopatias/veterinária , Animais , Biomarcadores/sangue , Bovinos , Doenças dos Bovinos/metabolismo , Indústria de Laticínios , Hepatopatias/sangue , Hepatopatias/metabolismo
16.
BMC Vet Res ; 10: 136, 2014 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-24946856

RESUMO

BACKGROUND: The influence of electromagnetic fields on the health of humans and animals is still an intensively discussed and scientifically investigated issue (Prakt Tierarzt 11:15-20, 2003; Umwelt Medizin Gesellschaft 17:326-332, 2004; J Toxicol Environment Health, Part B 12:572-597, 2009). We are surrounded by numerous electromagnetic fields of variable strength, coming from electronic equipment and its power cords, from high-voltage power lines and from antennas for radio, television and mobile communication. Particularly the latter cause's controversy, as everyone likes to have good mobile reception at anytime and anywhere, whereas nobody wants to have such a basestation antenna in their proximity. RESULTS: In this experiment, the NIR has resulted in changes in the enzyme activities. Certain enzymes were disabled, others enabled by NIR. Furthermore, individual behavior patterns were observed. While certain cows reacted to NIR, others did not react at all, or even inversely. CONCLUSION: The present results coincide with the information from the literature, according to which NIR leads to changes in redox proteins, and that there are individuals who are sensitive to radiation and others that are not. However, the latter could not be distinctly attributed - there are cows that react clearly with one enzyme while they do not react with another enzyme at all, or even the inverse. The study approach of testing ten cows each ten times during three phases has proven to be appropriate. Future studies should however set the post-exposure phase later on.


Assuntos
Catalase/metabolismo , Bovinos/sangue , Regulação Enzimológica da Expressão Gênica/efeitos da radiação , Glutationa Peroxidase/metabolismo , Radiação não Ionizante , Superóxido Dismutase/metabolismo , Animais , Catalase/sangue , Glutationa Peroxidase/sangue , Superóxido Dismutase/sangue
17.
Nat Commun ; 5: 3695, 2014 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-24770583

RESUMO

DNA damage recognition subunits such as DDB2 and XPC protect the human skin from ultraviolet (UV) light-induced genome instability and cancer, as demonstrated by the devastating inherited syndrome xeroderma pigmentosum. Here we show that the beneficial DNA repair response triggered by these two genome caretakers critically depends on a dynamic spatiotemporal regulation of their homeostasis. The prolonged retention of DDB2 and XPC in chromatin, because of a failure to readily remove both recognition subunits by the ubiquitin-dependent p97/VCP/Cdc48 segregase complex, leads to impaired DNA excision repair of UV lesions. Surprisingly, the ensuing chromosomal aberrations in p97-deficient cells are alleviated by a concomitant downregulation of DDB2 or XPC. Also, genome instability resulting from an excess of DDB2 persisting in UV-irradiated cells is prevented by concurrent p97 overexpression. Our findings demonstrate that DNA damage sensors and repair initiators acquire unexpected genotoxic properties if not controlled by timely extraction from chromatin.


Assuntos
Cromatina/metabolismo , Dano ao DNA/fisiologia , Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Clonagem Molecular , Ensaio de Unidades Formadoras de Colônias , Primers do DNA/genética , Instabilidade Genômica/genética , Instabilidade Genômica/fisiologia , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Microscopia de Fluorescência , RNA Interferente Pequeno/genética , Raios Ultravioleta
18.
PLoS One ; 9(4): e94149, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24722772

RESUMO

OBJECTIVE: Formaldehyde is still widely employed as a universal crosslinking agent, preservative and disinfectant, despite its proven carcinogenicity in occupationally exposed workers. Therefore, it is of paramount importance to understand the possible impact of low-dose formaldehyde exposures in the general population. Due to the concomitant occurrence of multiple indoor and outdoor toxicants, we tested how formaldehyde, at micromolar concentrations, interferes with general DNA damage recognition and excision processes that remove some of the most frequently inflicted DNA lesions. METHODOLOGY/PRINCIPAL FINDINGS: The overall mobility of the DNA damage sensors UV-DDB (ultraviolet-damaged DNA-binding) and XPC (xeroderma pigmentosum group C) was analyzed by assessing real-time protein dynamics in the nucleus of cultured human cells exposed to non-cytotoxic (<100 µM) formaldehyde concentrations. The DNA lesion-specific recruitment of these damage sensors was tested by monitoring their accumulation at local irradiation spots. DNA repair activity was determined in host-cell reactivation assays and, more directly, by measuring the excision of DNA lesions from chromosomes. Taken together, these assays demonstrated that formaldehyde obstructs the rapid nuclear trafficking of DNA damage sensors and, consequently, slows down their relocation to DNA damage sites thus delaying the excision repair of target lesions. A concentration-dependent effect relationship established a threshold concentration of as low as 25 micromolar for the inhibition of DNA excision repair. CONCLUSIONS/SIGNIFICANCE: A main implication of the retarded repair activity is that low-dose formaldehyde may exert an adjuvant role in carcinogenesis by impeding the excision of multiple mutagenic base lesions. In view of this generally disruptive effect on DNA repair, we propose that formaldehyde exposures in the general population should be further decreased to help reducing cancer risks.


Assuntos
Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Formaldeído/toxicidade , Transporte Ativo do Núcleo Celular , Células Cultivadas , Cromatina/química , Meios de Cultura , Proteínas de Ligação a DNA/genética , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Recuperação de Fluorescência Após Fotodegradação , Humanos , Raios Ultravioleta
19.
Curr Biol ; 23(3): 204-12, 2013 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-23352696

RESUMO

BACKGROUND: Nucleotide excision repair is a versatile DNA repair reaction that removes bulky adducts generated by environmental mutagens such as the UV spectrum of sunlight or chemical carcinogens. Current multistep models of this excision repair pathway accommodate its broad substrate repertoire but fail to explain the stringent selectivity toward damaged nucleotides among excess native DNA. To understand the mechanism of bulky lesion recognition, we postulated that it is necessary to analyze the function of xeroderma pigmentosum group D (XPD) protein beyond its well-known role in the unwinding of double-stranded DNA. RESULTS: We engineered two new XPD mutants (Y192A and R196E), involving amino acid substitutions near its central protein pore, that confer defective DNA repair despite normal transcription. In situ fluorescence-based protein dynamics studies in living cells demonstrated that both new mutants were unable to recognize DNA damage and failed to form stable associations with lesion sites. However, when their biochemical properties were tested in the framework of an archaeal protein homolog, they both retained ATPase and DNA-unwinding activity. The outstanding difference versus the wild-type control was that their directional 5'-3' translocation along DNA was not stopped by a bulky lesion, and moreover, they were unable to build long-lived demarcation complexes at damaged sites. CONCLUSIONS: By uncoupling for the first time the unwinding and damage sensor activities of XPD, we describe an unprecedented genome quality control process whereby a recognition pocket near the central DNA helicase pore scans individual substrate strands to capture base adducts.


Assuntos
Reparo do DNA , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo , Linhagem Celular Transformada , Núcleo Celular/metabolismo , DNA/metabolismo , Dano ao DNA , Humanos , Mutação
20.
PLoS Biol ; 9(10): e1001183, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22039351

RESUMO

How tightly packed chromatin is thoroughly inspected for DNA damage is one of the fundamental unanswered questions in biology. In particular, the effective excision of carcinogenic lesions caused by the ultraviolet (UV) radiation of sunlight depends on UV-damaged DNA-binding protein (UV-DDB), but the mechanism by which this DDB1-DDB2 heterodimer stimulates DNA repair remained enigmatic. We hypothesized that a distinctive function of this unique sensor is to coordinate damage recognition in the nucleosome repeat landscape of chromatin. Therefore, the nucleosomes of human cells have been dissected by micrococcal nuclease, thus revealing, to our knowledge for the first time, that UV-DDB associates preferentially with lesions in hypersensitive, hence, highly accessible internucleosomal sites joining the core particles. Surprisingly, the accompanying CUL4A ubiquitin ligase activity is necessary to retain the xeroderma pigmentosum group C (XPC) partner at such internucleosomal repair hotspots that undergo very fast excision kinetics. This CUL4A complex thereby counteracts an unexpected affinity of XPC for core particles that are less permissive than hypersensitive sites to downstream repair subunits. That UV-DDB also adopts a ubiquitin-independent function is evidenced by domain mapping and in situ protein dynamics studies, revealing direct but transient interactions that promote a thermodynamically unfavorable ß-hairpin insertion of XPC into substrate DNA. We conclude that the evolutionary advent of UV-DDB correlates with the need for a spatiotemporal organizer of XPC positioning in higher eukaryotic chromatin.


Assuntos
Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Nucleossomos/metabolismo , Animais , Benzoatos/farmacologia , Células CHO , Cricetinae , Proteínas Culina/genética , Proteínas Culina/metabolismo , DNA/metabolismo , DNA/efeitos da radiação , Proteínas de Ligação a DNA/genética , Furanos/farmacologia , Técnicas de Silenciamento de Genes , Células HEK293 , Células HeLa , Humanos , Ligação Proteica , Transporte Proteico , Pirazóis/farmacologia , Dímeros de Pirimidina/metabolismo , Interferência de RNA , Enzimas Ativadoras de Ubiquitina/antagonistas & inibidores , Ubiquitinação , Raios Ultravioleta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA