Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Hepatol ; 77(1): 71-83, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35131407

RESUMEN

BACKGROUND & AIMS: Acetaminophen (APAP) overdose remains a frequent cause of acute liver failure, which is generally accompanied by increased levels of serum bile acids (BAs). However, the pathophysiological role of BAs remains elusive. Herein, we investigated the role of BAs in APAP-induced hepatotoxicity. METHODS: We performed intravital imaging to investigate BA transport in mice, quantified endogenous BA concentrations in the serum of mice and patients with APAP overdose, analyzed liver tissue and bile by mass spectrometry and MALDI-mass spectrometry imaging, assessed the integrity of the blood-bile barrier and the role of oxidative stress by immunostaining of tight junction proteins and intravital imaging of fluorescent markers, identified the intracellular cytotoxic concentrations of BAs, and performed interventions to block BA uptake from blood into hepatocytes. RESULTS: Prior to the onset of cell death, APAP overdose causes massive oxidative stress in the pericentral lobular zone, which coincided with a breach of the blood-bile barrier. Consequently, BAs leak from the bile canaliculi into the sinusoidal blood, which is then followed by their uptake into hepatocytes via the basolateral membrane, their secretion into canaliculi and repeated cycling. This, what we termed 'futile cycling' of BAs, led to increased intracellular BA concentrations that were high enough to cause hepatocyte death. Importantly, however, the interruption of BA re-uptake by pharmacological NTCP blockage using Myrcludex B and Oatp knockout strongly reduced APAP-induced hepatotoxicity. CONCLUSIONS: APAP overdose induces a breach of the blood-bile barrier which leads to futile BA cycling that causes hepatocyte death. Prevention of BA cycling may represent a therapeutic option after APAP intoxication. LAY SUMMARY: Only one drug, N-acetylcysteine, is approved for the treatment of acetaminophen overdose and it is only effective when given within ∼8 hours after ingestion. We identified a mechanism by which acetaminophen overdose causes an increase in bile acid concentrations (to above toxic thresholds) in hepatocytes. Blocking this mechanism prevented acetaminophen-induced hepatotoxicity in mice and evidence from patients suggests that this therapy may be effective for longer periods after ingestion compared to N-acetylcysteine.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Sobredosis de Droga , Acetaminofén/metabolismo , Acetilcisteína/farmacología , Animales , Ácidos y Sales Biliares/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL
2.
Hepatology ; 74(2): 973-986, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33872408

RESUMEN

BACKGROUND AND AIMS: The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) regulates an array of cytoprotective genes, yet studies in transgenic mice have led to conflicting reports on its role in liver regeneration. We aimed to test the hypothesis that pharmacological activation of Nrf2 would enhance liver regeneration. APPROACH AND RESULTS: Wild-type and Nrf2 null mice were administered bardoxolone methyl (CDDO-Me), a potent activator of Nrf2 that has entered clinical development, and then subjected to two-thirds partial hepatectomy. Using translational noninvasive imaging techniques, CDDO-Me was shown to enhance the rate of restoration of liver volume (MRI) and improve liver function (multispectral optoacoustic imaging of indocyanine green clearance) in wild-type, but not Nrf2 null, mice following partial hepatectomy. Using immunofluorescence imaging and whole transcriptome analysis, these effects were found to be associated with an increase in hepatocyte hypertrophy and proliferation, the suppression of immune and inflammatory signals, and metabolic adaptation in the remnant liver tissue. Similar processes were modulated following exposure of primary human hepatocytes to CDDO-Me, highlighting the potential relevance of our findings to patients. CONCLUSIONS: Our results indicate that pharmacological activation of Nrf2 is a promising strategy for enhancing functional liver regeneration. Such an approach could therefore aid the recovery of patients undergoing liver surgery and support the treatment of acute and chronic liver disease.


Asunto(s)
Regeneración Hepática/efectos de los fármacos , Hígado/efectos de los fármacos , Factor 2 Relacionado con NF-E2/agonistas , Ácido Oleanólico/análogos & derivados , Adulto , Anciano de 80 o más Años , Animales , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hepatectomía , Hepatocitos , Humanos , Hígado/fisiología , Hígado/cirugía , Regeneración Hepática/genética , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Ácido Oleanólico/administración & dosificación , Cultivo Primario de Células
3.
Arch Toxicol ; 93(2): 385-399, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30426165

RESUMEN

The transcription factor NRF2, governed by its repressor KEAP1, protects cells against oxidative stress. There is interest in modelling the NRF2 response to improve the prediction of clinical toxicities such as drug-induced liver injury (DILI). However, very little is known about the makeup of the NRF2 transcriptional network and its response to chemical perturbation in primary human hepatocytes (PHH), which are often used as a translational model for investigating DILI. Here, microarray analysis identified 108 transcripts (including several putative novel NRF2-regulated genes) that were both downregulated by siRNA targeting NRF2 and upregulated by siRNA targeting KEAP1 in PHH. Applying weighted gene co-expression network analysis (WGCNA) to transcriptomic data from the Open TG-GATES toxicogenomics repository (representing PHH exposed to 158 compounds) revealed four co-expressed gene sets or 'modules' enriched for these and other NRF2-associated genes. By classifying the 158 TG-GATES compounds based on published evidence, and employing the four modules as network perturbation metrics, we found that the activation of NRF2 is a very good indicator of the intrinsic biochemical reactivity of a compound (i.e. its propensity to cause direct chemical stress), with relatively high sensitivity, specificity, accuracy and positive/negative predictive values. We also found that NRF2 activation has lower sensitivity for the prediction of clinical DILI risk, although relatively high specificity and positive predictive values indicate that false positive detection rates are likely to be low in this setting. Underpinned by our comprehensive analysis, activation of the NRF2 network is one of several mechanism-based components that can be incorporated into holistic systems toxicology models to improve mechanistic understanding and preclinical prediction of DILI in man.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Redes Reguladoras de Genes/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Factor 2 Relacionado con NF-E2/genética , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/patología , Humanos , Isotiocianatos/efectos adversos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , ARN Interferente Pequeño , Sulfóxidos
5.
Kidney Int ; 88(6): 1261-1273, 2015 12.
Artículo en Inglés | MEDLINE | ID: mdl-26422507

RESUMEN

The transcription factor Nrf2 exerts protective effects in numerous experimental models of acute kidney injury, and is a promising therapeutic target in chronic kidney disease. To provide a detailed insight into the regulatory roles of Nrf2 in the kidney, we performed integrated transcriptomic and proteomic analyses of kidney tissue from wild-type and Nrf2 knockout mice treated with the Nrf2 inducer methyl-2-cyano-3,12-dioxooleano-1,9-dien-28-oate (CDDO-Me, also known as bardoxolone methyl). After 24 h, analyses identified 2561 transcripts and 240 proteins that were differentially expressed in the kidneys of Nrf2 knockout mice, compared with those of wild-type counterparts, and 3122 transcripts and 68 proteins that were differentially expressed in wild-type mice treated with CDDO-Me, compared with those of vehicle control. In the light of their sensitivity to genetic and pharmacological modulation of renal Nrf2 activity, genes/proteins that regulate xenobiotic disposition, redox balance, the intra/extracellular transport of small molecules, and the supply of NADPH and other cellular fuels were found to be positively regulated by Nrf2 in the kidney. This was verified by qPCR, immunoblotting, pathway analysis, and immunohistochemistry. In addition, the levels of NADPH and glutathione were found to be significantly decreased in the kidneys of Nrf2 knockout mice. Thus, Nrf2 regulates genes that coordinate homeostatic processes in the kidney, highlighting its potential as a novel therapeutic target.

6.
Biochem Soc Trans ; 43(4): 657-62, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26551708

RESUMEN

Beyond specific limits of exposure, chemical entities can provoke deleterious effects in mammalian cells via direct interaction with critical macromolecules or by stimulating the accumulation of reactive oxygen species (ROS). In particular, these chemical and oxidative stresses can underpin adverse reactions to therapeutic drugs, which pose an unnecessary burden in the clinic and pharmaceutical industry. Novel pre-clinical testing strategies are required to identify, at an earlier stage in the development pathway, chemicals and drugs that are likely to provoke toxicity in humans. Mammalian cells can adapt to chemical and oxidative stress via the action of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), which up-regulates the expression of numerous cell defence genes and has been shown to protect against a variety of chemical toxicities. Here, we provide a brief overview of the Nrf2 pathway and summarize novel experimental models that can be used to monitor changes in Nrf2 pathway activity and thus understand the functional consequences of such perturbations in the context of chemical and drug toxicity. We also provide an outlook on the potential value of monitoring Nrf2 activity for improving the pre-clinical identification of chemicals and drugs with toxic liability in humans.


Asunto(s)
Sustancias Macromoleculares/toxicidad , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
7.
Redox Biol ; 72: 103134, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38643749

RESUMEN

The cytoprotective transcription factor NRF2 regulates the expression of several hundred genes in mammalian cells and is a promising therapeutic target in a number of diseases associated with oxidative stress and inflammation. Hence, an ability to monitor basal and inducible NRF2 signalling is vital for mechanistic understanding in translational studies. Due to some caveats related to the direct measurement of NRF2 levels, the modulation of NRF2 activity is typically determined by measuring changes in the expression of one or more of its target genes and/or the associated protein products. However, there is a lack of consensus regarding the most relevant set of these genes/proteins that best represents NRF2 activity across cell types and species. We present the findings of a comprehensive literature search that according to stringent criteria identifies GCLC, GCLM, HMOX1, NQO1, SRXN1 and TXNRD1 as a robust panel of markers that are directly regulated by NRF2 in multiple cell and tissue types. We assess the relevance of these markers in clinically accessible biofluids and highlight future challenges in the development and use of NRF2 biomarkers in humans.


Asunto(s)
Biomarcadores , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Transducción de Señal , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Humanos , Animales , Regulación de la Expresión Génica
8.
Kidney Int ; 84(6): 1090-5, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23783243

RESUMEN

Multifaceted cell defense pathways perform a critical role in the maintenance of homeostasis at the cellular, tissue, and organism levels. The Keap1-Nrf2 pathway is one of the most important of these cytoprotective pathways, with Nrf2 serving as a master transcriptional regulator of the basal and inducible expression of a multitude of genes encoding detoxification enzymes, antioxidant proteins, xenobiotic transporters, and other stress-response mediators. An increasing body of evidence supports a vital physiological role for Nrf2 in protection of the kidney against a number of diseases, and the pharmacological induction of Nrf2 by bardoxolone methyl (methyl-2-cyano 3,12-dioxooleano-1,9-dien-28-oate, CDDO-Me) has shown promise for the management of such pathologies. Acute kidney injury, induced by drugs and other stimuli, is a significant clinical problem, and accounts for the cessation of development of many promising drug candidates. A better understanding of the molecular mechanisms that underlie acute kidney injury, and the biological facets that determine the balance between renal adaptation and dysfunction, is therefore vital to reducing clinical burden and patient suffering. The focus of this review is to highlight recent work that has demonstrated an ability of Nrf2 to determine the sensitivity of the kidney to acute injury invoked by environmental insults such as heavy metals and ischemia, as well as xenobiotics such as cyclosporin A and cisplatin.


Asunto(s)
Lesión Renal Aguda/prevención & control , Riñón/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Agentes Urológicos/uso terapéutico , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Animales , Citoprotección , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
9.
Mol Ther Nucleic Acids ; 31: 211-223, 2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36700046

RESUMEN

Tumor suppressor genes (TSGs) are frequently downregulated in cancer, leading to dysregulation of the pathways that they control. The continuum model of tumor suppression suggests that even subtle changes in TSG expression, for example, driven by epigenetic modifications or copy number alterations, can lead to a loss of gene function and a phenotypic effect. This approach to exploring tumor suppression provides opportunities for alternative therapies that may be able to restore TSG expression toward normal levels, such as oligonucleotide therapies. Oligonucleotide therapies involve the administration of exogenous nucleic acids to modulate the expression of specific endogenous genes. This review focuses on two types of activating oligonucleotide therapies, small-activating RNAs and synthetic mRNAs, as novel methods to increase the expression of TSGs in cancer.

10.
Trends Pharmacol Sci ; 44(3): 137-149, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36628798

RESUMEN

Activation of the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) is emerging as an attractive therapeutic approach to counteract oxidative stress, inflammation, and metabolic imbalances. These processes underpin many chronic pathologies with unmet therapeutic needs, including neurodegenerative disorders and metabolic diseases. As the NRF2 field transitions into the clinical phase of its evolution, the need for an understanding of the factors influencing NRF2 pharmacology has never been greater. In this opinion article we describe the rationale for targeting NRF2, summarise the recent advances in drug development of NRF2 modulators, and reflect on the remaining challenges in realising the full clinical potential of NRF2 as a therapeutic target.


Asunto(s)
Terapia Molecular Dirigida , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Humanos , Desarrollo de Medicamentos , Inflamación/tratamiento farmacológico , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos
11.
Toxicol Sci ; 196(1): 112-125, 2023 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-37647630

RESUMEN

To minimize the occurrence of unexpected toxicities in early phase preclinical studies of new drugs, it is vital to understand fundamental similarities and differences between preclinical species and humans. Species differences in sensitivity to acetaminophen (APAP) liver injury have been related to differences in the fraction of the drug that is bioactivated to the reactive metabolite N-acetyl-p-benzoquinoneimine (NAPQI). We have used physiologically based pharmacokinetic modeling to identify oral doses of APAP (300 and 1000 mg/kg in mice and rats, respectively) yielding similar hepatic burdens of NAPQI to enable the comparison of temporal liver tissue responses under conditions of equivalent chemical insult. Despite pharmacokinetic and biochemical verification of the equivalent NAPQI insult, serum biomarker and tissue histopathology analyses revealed that mice still exhibited a greater degree of liver injury than rats. Transcriptomic and proteomic analyses highlighted the stronger activation of stress response pathways (including the Nrf2 oxidative stress response and autophagy) in the livers of rats, indicative of a more robust transcriptional adaptation to the equivalent insult. Components of these pathways were also found to be expressed at a higher basal level in the livers of rats compared with both mice and humans. Our findings exemplify a systems approach to understanding differential species sensitivity to hepatotoxicity. Multiomics analysis indicated that rats possess a greater basal and adaptive capacity for hepatic stress responses than mice and humans, with important implications for species selection and human translation in the safety testing of new drug candidates associated with reactive metabolite formation.


Asunto(s)
Acetaminofén , Enfermedad Hepática Inducida por Sustancias y Drogas , Ratas , Ratones , Humanos , Animales , Acetaminofén/toxicidad , Acetaminofén/metabolismo , Proteómica , Especificidad de la Especie , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Hígado/metabolismo , Estrés Oxidativo , Análisis de Sistemas
12.
J Biol Chem ; 286(2): 987-96, 2011 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-21059641

RESUMEN

The artemisinin compounds are the frontline drugs for the treatment of drug-resistant malaria. They are selectively cytotoxic to mammalian cancer cell lines and have been implicated as neurotoxic and embryotoxic in animal studies. The endoperoxide functional group is both the pharmacophore and toxicophore, but the proposed chemical mechanisms and targets of cytotoxicity remain unclear. In this study we have used cell models and quantitative drug metabolite analysis to define the role of the mitochondrion and cellular heme in the chemical and molecular mechanisms of cell death induced by artemisinin compounds. HeLa ρ(0) cells, which are devoid of a functioning electron transport chain, were used to demonstrate that actively respiring mitochondria play an essential role in endoperoxide-induced cytotoxicity (artesunate IC(50) values, 48 h: HeLa cells, 6 ± 3 µM; and HeLa ρ(0) cells, 34 ± 5 µM) via the generation of reactive oxygen species and the induction of mitochondrial dysfunction and apoptosis but do not have any role in the reductive activation of the endoperoxide to cytotoxic carbon-centered radicals. However, using chemical modulators of heme synthesis (succinylacetone and protoporphyrin IX) and cellular iron content (holotransferrin), we have demonstrated definitively that free or protein-bound heme is responsible for intracellular activation of the endoperoxide group and that this is the chemical basis of cytotoxicity (IC(50) value and biomarker of bioactivation levels, respectively: 10ß-(p-fluorophenoxy)dihydroartemisinin alone, 0.36 ± 0.20 µM and 11 ± 5%; and with succinylacetone, >100 µM and 2 ± 5%).


Asunto(s)
Antimaláricos/toxicidad , Apoptosis/efectos de los fármacos , Artemisininas/toxicidad , Hemo/metabolismo , Mitocondrias/efectos de los fármacos , Antimaláricos/química , Apoptosis/fisiología , Artemisininas/química , Artesunato , Células HeLa , Humanos , Hierro/metabolismo , Mitocondrias/metabolismo , Peróxidos/metabolismo , Especies Reactivas de Oxígeno/metabolismo
13.
Mol Med ; 18: 1045-55, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22669474

RESUMEN

Semisynthetic artemisinin-based therapies are the first-line treatment for P. falciparum malaria, but next-generation synthetic drug candidates are urgently required to improve availability and respond to the emergence of artemisinin-resistant parasites. Artemisinins are embryotoxic in animal models and induce apoptosis in sensitive mammalian cells. Understanding the cytotoxic propensities of antimalarial drug candidates is crucial to their successful development and utilization. Here, we demonstrate that, similarly to the model artemisinin artesunate (ARS), a synthetic tetraoxane drug candidate (RKA182) and a trioxolane equivalent (FBEG100) induce embryotoxicity and depletion of primitive erythroblasts in a rodent model. We also show that RKA182, FBEG100 and ARS are cytotoxic toward a panel of established and primary human cell lines, with caspase-dependent apoptosis and caspase-independent necrosis underlying the induction of cell death. Although the toxic effects of RKA182 and FBEG100 proceed more rapidly and are relatively less cell-selective than that of ARS, all three compounds are shown to be dependent upon heme, iron and oxidative stress for their ability to induce cell death. However, in contrast to previously studied artemisinins, the toxicity of RKA182 and FBEG100 is shown to be independent of general chemical decomposition. Although tetraoxanes and trioxolanes have shown promise as next-generation antimalarials, the data described here indicate that adverse effects associated with artemisinins, including embryotoxicity, cannot be ruled out with these novel compounds, and a full understanding of their toxicological actions will be central to the continuing design and development of safe and effective drug candidates which could prove important in the fight against malaria.


Asunto(s)
Antimaláricos/toxicidad , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/patología , Teratógenos/toxicidad , Tetraoxanos/toxicidad , Animales , Antimaláricos/química , Artemisininas/química , Artemisininas/toxicidad , Artesunato , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular , Femenino , Hemo/metabolismo , Humanos , Hierro/metabolismo , Masculino , Estrés Oxidativo/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Compuestos de Espiro/química , Compuestos de Espiro/toxicidad , Tetraoxanos/química , Factores de Tiempo
14.
J Biol Chem ; 285(22): 16782-8, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20378532

RESUMEN

Nrf2 regulates the expression of numerous cytoprotective genes in mammalian cells. The activity of Nrf2 is regulated by the Cul3 adaptor Keap1, yet little is known regarding mechanisms of regulation of Keap1 itself. Here, we have used immunopurification of Keap1 and mass spectrometry, in addition to immunoblotting, to identify sequestosome 1 (SQSTM1) as a cellular binding partner of Keap1. SQSTM1 serves as a scaffold in various signaling pathways and shuttles polyubiquitinated proteins to the proteasomal and lysosomal degradation machineries. Ectopic expression of SQSTM1 led to a decrease in the basal protein level of Keap1 in a panel of cells. Furthermore, RNA interference (RNAi) depletion of SQSTM1 resulted in an increase in the protein level of Keap1 and a concomitant decrease in the protein level of Nrf2 in the absence of changes in Keap1 or Nrf2 mRNA levels. The increased protein level of Keap1 in cells depleted of SQSTM1 by RNAi was linked to a decrease in its rate of degradation; the half-life of Keap1 was almost doubled by RNAi depletion of SQSTM1. The decreased level of Nrf2 in cells depleted of SQSTM1 by RNAi was associated with decreases in the mRNA levels, protein levels, and function of several Nrf2-regulated cell defense genes. SQSTM1 was dispensable for the induction of the Keap1-Nrf2 pathway, as Nrf2 activation by tert-butylhydroquinone or iodoacetamide was not affected by RNAi depletion of SQSTM1. These findings demonstrate a physical and functional interaction between Keap1 and SQSTM1 and reveal an additional layer of regulation in the Keap1-Nrf2 pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/fisiología , Factor 2 Relacionado con NF-E2/fisiología , Animales , Línea Celular , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch , Lisosomas/metabolismo , Ratones , Modelos Biológicos , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Interferencia de ARN , Proteína Sequestosoma-1 , Transducción de Señal
15.
Mol Cancer ; 10: 37, 2011 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-21489257

RESUMEN

BACKGROUND: Nrf2 is a key transcriptional regulator of a battery of genes that facilitate phase II/III drug metabolism and defence against oxidative stress. Nrf2 is largely regulated by Keap1, which directs Nrf2 for proteasomal degradation. The Nrf2/Keap1 system is dysregulated in lung, head and neck, and breast cancers and this affects cellular proliferation and response to therapy. Here, we have investigated the integrity of the Nrf2/Keap1 system in pancreatic cancer. RESULTS: Keap1, Nrf2 and the Nrf2 target genes AKR1c1 and GCLC were detected in a panel of five pancreatic cancer cell lines. Mutation analysis of NRF2 exon 2 and KEAP1 exons 2-6 in these cell lines identified no mutations in NRF2 and only synonomous mutations in KEAP1. RNAi depletion of Nrf2 caused a decrease in the proliferation of Suit-2, MiaPaca-2 and FAMPAC cells and enhanced sensitivity to gemcitabine (Suit-2), 5-flurouracil (FAMPAC), cisplatin (Suit-2 and FAMPAC) and gamma radiation (Suit-2). The expression of Nrf2 and Keap1 was also analysed in pancreatic ductal adenocarcinomas (n = 66 and 57, respectively) and matching normal benign epithelium (n = 21 cases). Whilst no significant correlation was seen between the expression levels of Keap1 and Nrf2 in the tumors, interestingly, Nrf2 staining was significantly greater in the cytoplasm of tumors compared to benign ducts (P < 0.001). CONCLUSIONS: Expression of Nrf2 is up-regulated in pancreatic cancer cell lines and ductal adenocarcinomas. This may reflect a greater intrinsic capacity of these cells to respond to stress signals and resist chemotherapeutic interventions. Nrf2 also appears to support proliferation in certain pancreatic adenocarinomas. Therefore, strategies to pharmacologically manipulate the levels and/or activity of Nrf2 may have the potential to reduce pancreatic tumor growth, and increase sensitivity to therapeutics.


Asunto(s)
Proliferación Celular , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias Pancreáticas/genética , Línea Celular Tumoral , Exones , Regulación Neoplásica de la Expresión Génica , Genotipo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch , Factor 2 Relacionado con NF-E2/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Transducción de Señal , Regulación hacia Arriba
16.
Handb Exp Pharmacol ; (196): 233-66, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20020265

RESUMEN

Adverse drug reactions pose a significant public health problem. In some cases, the process of drug metabolism can contribute to the onset of toxicity through the bioactivation of a parent molecule to a chemically reactive intermediate. In order to maintain a favorable balance between bioactivation and detoxification, mammalian cells have evolved an inducible cell defense system known as the antioxidant response pathway. The activity of this cytoprotective pathway is largely regulated by the transcription factor Nrf2, which governs the expression of many phase II detoxification and antioxidant enzymes. In turn, the activity of Nrf2 is regulated by the cysteine-rich cytosolic inhibitor Keap1, which acts as a "sensor" for chemical/oxidative stress. This article summarizes our current understanding of the molecular mechanisms that regulate the function of the Keap1-Nrf2 pathway and highlights the importance of Nrf2 in the protection against drug-induced toxicity.


Asunto(s)
Hipersensibilidad a las Drogas/prevención & control , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Antioxidantes/metabolismo , Biotransformación , Citoprotección , Hipersensibilidad a las Drogas/etiología , Hipersensibilidad a las Drogas/genética , Hipersensibilidad a las Drogas/metabolismo , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína 1 Asociada A ECH Tipo Kelch , Factor 2 Relacionado con NF-E2/genética , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Polimorfismo Genético , Transducción de Señal/genética , Ubiquitinación
17.
Nat Rev Drug Discov ; 19(2): 131-148, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31748707

RESUMEN

Drug-induced liver injury (DILI) is a patient-specific, temporal, multifactorial pathophysiological process that cannot yet be recapitulated in a single in vitro model. Current preclinical testing regimes for the detection of human DILI thus remain inadequate. A systematic and concerted research effort is required to address the deficiencies in current models and to present a defined approach towards the development of new or adapted model systems for DILI prediction. This Perspective defines the current status of available models and the mechanistic understanding of DILI, and proposes our vision of a roadmap for the development of predictive preclinical models of human DILI.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/diagnóstico , Modelos Animales de Enfermedad , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Humanos , Valor Predictivo de las Pruebas
18.
Hepatology ; 48(4): 1292-301, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18785192

RESUMEN

UNLABELLED: The transcription factor Nrf2 regulates the expression of numerous cytoprotective genes in mammalian cells. We have demonstrated previously that acetaminophen activates Nrf2 in mouse liver following administration of non-hepatotoxic and hepatotoxic doses in vivo, implying that Nrf2 may have an important role in the protection against drug-induced liver injury. Nrf2 activation has been proposed to occur through the modification of cysteine residues within Keap1, the cytosolic repressor of Nrf2. We hypothesized that acetaminophen activates Nrf2 via the formation of its reactive metabolite N-acetyl-p-benzoquinoneimine (NAPQI), which may disrupt the repression of Nrf2 through the modification of cysteine residues within Keap1. Here, we show that NAPQI can directly activate the Nrf2 pathway in mouse liver cells, inducing an adaptive defense response that is antagonized by RNA interference targeted against Nrf2. Furthermore, mass spectrometric analysis shows that NAPQI selectively modifies cysteine residues in Keap1, both in recombinant protein in vitro and in cells ectopically expressing Keap1. Using this cell-based model, we demonstrate that activation of Nrf2 by NAPQI and a panel of probe molecules [dexamethasone 21-mesylate, 15-deoxy-Delta-((12,14))-prostaglandin J(2), 2,4-dinitrochlorobenzene, and iodoacetamide] correlates with the selective modification of cysteine residues located within the intervening region of Keap1. However, substantial depletion of glutathione (to less than 15% of basal levels) by buthionine sulfoximine, which does not directly modify Keap1, is also sufficient to activate Nrf2. CONCLUSION: Nrf2 can be activated via the direct modification of cysteine residues located within the intervening region of Keap1, but also via the substantial depletion of glutathione without the requirement for direct modification of Keap1. It is possible that both of these mechanisms contribute to the activation of Nrf2 by acetaminophen.


Asunto(s)
Acetaminofén/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Benzoquinonas/farmacología , Proteínas del Citoesqueleto/metabolismo , Iminas/farmacología , Hígado/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Acetaminofén/farmacología , Proteínas Adaptadoras Transductoras de Señales/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular , Línea Celular Tumoral , Proteínas del Citoesqueleto/efectos de los fármacos , Proteínas del Citoesqueleto/genética , Glutatión/metabolismo , Humanos , Proteína 1 Asociada A ECH Tipo Kelch , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Factor 2 Relacionado con NF-E2/genética , Interferencia de ARN , Transducción de Señal/fisiología
19.
Mult Scler Relat Disord ; 30: 257-261, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30851639

RESUMEN

BACKGROUND: Nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2) is a master transcriptional regulator of the protective cellular response to oxidative stress. Sulforaphane is a Nrf2 activator but is unstable at ambient temperature. SFX-01 is a novel composition comprised of synthetic sulforaphane stabilised within the pocket of an α-cyclodextrin complex. Here we tested the efficacy of SFX-01 in murine relapsing experimental autoimmune encephalomyelitis (EAE), a model of relapsing-remitting MS (RRMS). METHODS: Relapsing EAE was induced in female SJL mice using immunization against PLP139-151. In the therapeutic experiment, the aim was to model initiation of treatment after diagnosis in RRMS, so treatment was started at day 19, one day prior to the expected relapse onset. In the prophylactic experiment, mice were treated from the time of immunization and followed for three weeks. RESULTS: SFX-01 reduced residual disability in both experiments. Most of this effect was mediated by a decrease in maximum severity of relapses and improved recovery during follow-up. Histological examination of the spinal cord was consistent with the clinical findings, with improvement in demyelination and the number of apoptotic cells, but not inflammatory cell infiltration, compared to the vehicle group. CONCLUSIONS: SFX-01 is efficacious in EAE. In first-in-man and phase II clinical trials for other indications, SFX-01 was found to be well-tolerated. A trial comparing BG-12 and SFX-01 would address whether SFX-01 can offer RRMS patients a better option with respect to efficacy and tolerability.


Asunto(s)
Anticarcinógenos/uso terapéutico , Encefalomielitis Autoinmune Experimental/complicaciones , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Isotiocianatos/uso terapéutico , Resultado del Tratamiento , Animales , Evaluación de la Discapacidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Encefalomielitis Autoinmune Experimental/inducido químicamente , Femenino , Adyuvante de Freund/toxicidad , Estimación de Kaplan-Meier , Ratones , Proteína Proteolipídica de la Mielina/toxicidad , Fragmentos de Péptidos/toxicidad , Sulfóxidos , Factores de Tiempo
20.
Biomed Pharmacother ; 112: 108637, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30798127

RESUMEN

Dose-dependent cardiotoxicity is the leading adverse reaction seen in cancer patients treated with doxorubicin. Currently, dexrazoxane is the only approved drug that can partially protect against this toxicity in patients, however, its administration is restricted to those patients receiving a high cumulative dose of anthracyclines. Investigations into the mechanisms of cardiotoxicity and efforts to improve cardioprotective strategies have been hindered by the limited availability of a phenotypically relevant in vitro adult human cardiac model system. Here, we adapted a readily reproducible, functional 3D human multi-cell type cardiac system to emulate patient responses seen with doxorubicin and dexrazoxane. We show that administration of two NRF2 gene inducers namely the semi-synthetic triterpenoid Bardoxolone methyl, and the isothiocyanate sulfurophane, result in cardioprotection against doxorubicin toxicity comparable to dexrazoxane as evidenced by an increase in cell viability and a decrease in the production of reactive oxygen species. We further show a synergistic attenuation of cardiotoxicity when the NRF2 inducers and dexrazoxane are used in tandem. Taken together, our data indicate that the 3D spheroid is a suitable model to investigate drug induced cardiotoxicity and we reveal an essential role of the NRF2 pathway in cardioprotection providing a novel pharmacological mechanism and intervention route towards the alleviation of doxorubicin-induced toxicity.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Doxorrubicina/toxicidad , Corazón/efectos de los fármacos , Factor 2 Relacionado con NF-E2/biosíntesis , Esferoides Celulares/efectos de los fármacos , Cardiotoxicidad/prevención & control , Supervivencia Celular/efectos de los fármacos , Dexrazoxano/farmacología , Sinergismo Farmacológico , Humanos , Técnicas In Vitro , Isotiocianatos/farmacología , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/farmacología , Especies Reactivas de Oxígeno/metabolismo , Esferoides Celulares/metabolismo , Sulfóxidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA