Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nature ; 569(7757): 570-575, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31019297

RESUMEN

Precision oncology hinges on linking tumour genotype with molecularly targeted drugs1; however, targeting the frequently dysregulated metabolic landscape of cancer has proven to be a major challenge2. Here we show that tissue context is the major determinant of dependence on the nicotinamide adenine dinucleotide (NAD) metabolic pathway in cancer. By analysing more than 7,000 tumours and 2,600 matched normal samples of 19 tissue types, coupled with mathematical modelling and extensive in vitro and in vivo analyses, we identify a simple and actionable set of 'rules'. If the rate-limiting enzyme of de novo NAD synthesis, NAPRT, is highly expressed in a normal tissue type, cancers that arise from that tissue will have a high frequency of NAPRT amplification and be completely and irreversibly dependent on NAPRT for survival. By contrast, tumours that arise from normal tissues that do not express NAPRT highly are entirely dependent on the NAD salvage pathway for survival. We identify the previously unknown enhancer that underlies this dependence. Amplification of NAPRT is shown to generate a pharmacologically actionable tumour cell dependence for survival. Dependence on another rate-limiting enzyme of the NAD synthesis pathway, NAMPT, as a result of enhancer remodelling is subject to resistance by NMRK1-dependent synthesis of NAD. These results identify a central role for tissue context in determining the choice of NAD biosynthetic pathway, explain the failure of NAMPT inhibitors, and pave the way for more effective treatments.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Amplificación de Genes , NAD/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Animales , Ligasas de Carbono-Nitrógeno con Glutamina como Donante de Amida-N/metabolismo , Muerte Celular , Línea Celular Tumoral , Citocinas/antagonistas & inhibidores , Citocinas/genética , Citocinas/metabolismo , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Neoplasias/enzimología , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Nicotinamida Fosforribosiltransferasa/genética , Nicotinamida Fosforribosiltransferasa/metabolismo , Pentosiltransferasa/genética , Pentosiltransferasa/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo
2.
Biochem Soc Trans ; 43(4): 611-20, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26551701

RESUMEN

Nuclear factor-erythroid 2 p45 (NF-E2 p45)-related factor 2 (Nrf2) is a master regulator of redox homoeostasis that allows cells to adapt to oxidative stress and also promotes cell proliferation. In this review, we describe the molecular mechanisms by which oxidants/electrophilic agents and growth factors increase Nrf2 activity. In the former case, oxidants/electrophiles increase the stability of Nrf2 by antagonizing the ability of Kelch-like ECH-associated protein 1 (Keap1) to target the transcription factor for proteasomal degradation via the cullin-3 (Cul3)-RING ubiquitin ligase CRL(Keap1). In the latter case, we speculate that growth factors increase the stability of Nrf2 by stimulating phosphoinositide 3-kinase (PI3K)-protein kinase B (PKB)/Akt signalling, which in turn results in inhibitory phosphorylation of glycogen synthase kinase-3 (GSK-3) and in doing so prevents the formation of a DSGIS motif-containing phosphodegron in Nrf2 that is recognized by the ß-transducin repeat-containing protein (ß-TrCP) Cul1-based E3 ubiquitin ligase complex SCF(ß-TrCP). We present data showing that in the absence of Keap1, the electrophile tert-butyl hydroquinone (tBHQ) can stimulate Nrf2 activity and induce the Nrf2-target gene NAD(P)H: quinone oxidoreductase-1 (NQO1), whilst simultaneously causing inhibitory phosphorylation of GSK-3ß at Ser(9). Together, these observations suggest that tBHQ can suppress the ability of SCF(ß-TrCP) to target Nrf2 for proteasomal degradation by increasing PI3K-PKB/Akt signalling. We also propose a scheme that explains how other protein kinases that inhibit GSK-3 could stimulate induction of Nrf2-target genes by preventing formation of the DSGIS motif-containing phosphodegron in Nrf2.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas con Repetición de beta-Transducina/metabolismo , Antioxidantes/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteína 1 Asociada A ECH Tipo Kelch , Factor 2 Relacionado con NF-E2/química , Estrés Oxidativo , Estabilidad Proteica , Estructura Terciaria de Proteína , Transducción de Señal/efectos de los fármacos
3.
Cancer Discov ; 13(1): 41-55, 2023 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-36355783

RESUMEN

With the combination of KRASG12C and EGFR inhibitors, KRAS is becoming a druggable target in colorectal cancer. However, secondary resistance limits its efficacy. Using cell lines, patient-derived xenografts, and patient samples, we detected a heterogeneous pattern of putative resistance alterations expected primarily to prevent inhibition of ERK signaling by drugs at progression. Serial analysis of patient blood samples on treatment demonstrates that most of these alterations are detected at a low frequency except for KRASG12C amplification, a recurrent resistance mechanism that rises in step with clinical progression. Upon drug withdrawal, resistant cells with KRASG12C amplification undergo oncogene-induced senescence, and progressing patients experience a rapid fall in levels of this alteration in circulating DNA. In this new state, drug resumption is ineffective as mTOR signaling is elevated. However, our work exposes a potential therapeutic vulnerability, whereby therapies that target the senescence response may overcome acquired resistance. SIGNIFICANCE: Clinical resistance to KRASG12C-EGFR inhibition primarily prevents suppression of ERK signaling. Most resistance mechanisms are subclonal, whereas KRASG12C amplification rises over time to drive a higher portion of resistance. This recurrent resistance mechanism leads to oncogene-induced senescence upon drug withdrawal and creates a potential vulnerability to senolytic approaches. This article is highlighted in the In This Issue feature, p. 1.


Asunto(s)
Neoplasias Colorrectales , Resistencia a Antineoplásicos , Animales , Humanos , Resistencia a Antineoplásicos/genética , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Transducción de Señal , Modelos Animales de Enfermedad , Receptores ErbB , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Mutación
5.
Redox Biol ; 47: 102158, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34626892

RESUMEN

The transcription factor Nrf2 is a stress-responsive master regulator of antioxidant, detoxification and proteostasis genes. In astrocytes, Nrf2-dependent gene expression drives cell-autonomous cytoprotection and also non-cell-autonomous protection of nearby neurons, and can ameliorate pathology in several acute and chronic neurological disorders associated with oxidative stress. However, the value of astrocytic Nrf2 as a therapeutic target depends in part on whether Nrf2 activation by disease-associated oxidative stress occludes the effect of any Nrf2-activating drug. Nrf2 activation classically involves the inhibition of interactions between Nrf2's Neh2 domain and Keap1, which directs Nrf2 degradation. Keap1 inhibition is mediated by the modification of cysteine residues on Keap1, and can be triggered by electrophilic small molecules such as tBHQ. Here we show that astrocytic Nrf2 activation by oxidative stress involves Keap1-independent non-canonical signaling. Keap1 deficiency elevates basal Nrf2 target gene expression in astrocytes and occludes the effects of tBHQ, oxidative stress still induced strong Nrf2-dependent gene expression in Keap1-deficient astrocytes. Moreover, while tBHQ prevented protein degradation mediated via Nrf2's Neh2 domain, oxidative stress did not, consistent with a Keap1-independent mechanism. Moreover the effects of oxidative stress and tBHQ on Nrf2 target gene expression are additive, not occlusive. Mechanistically, oxidative stress enhances the transactivation potential of Nrf2's Neh5 domain in a manner dependent on its Cys-191 residue. Thus, astrocytic Nrf2 activation by oxidative stress involves Keap1-independent non-canonical signaling, meaning that further Nrf2 activation by Keap1-inhibiting drugs may be a viable therapeutic strategy.


Asunto(s)
Astrocitos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Antioxidantes , Astrocitos/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/genética , Ratones , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo
6.
Nat Rev Cancer ; 20(1): 57-70, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31806884

RESUMEN

Altered cellular metabolism is a hallmark of gliomas. Propelled by a set of recent technological advances, new insights into the molecular mechanisms underlying glioma metabolism are rapidly emerging. In this Review, we focus on the dynamic nature of glioma metabolism and how it is shaped by the interaction between tumour genotype and brain microenvironment. Recent advances integrating metabolomics with genomics are discussed, yielding new insight into the mechanisms that drive glioma pathogenesis. Studies that shed light on interactions between the tumour microenvironment and tumour genotype are highlighted, providing important clues as to how gliomas respond to and adapt to their changing tissue and biochemical contexts. Finally, a road map for the discovery of potential new glioma drug targets is suggested, with the goal of translating these new insights about glioma metabolism into clinical benefits for patients.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Metabolismo Energético , Glioma/metabolismo , Animales , Biomarcadores , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/etiología , Neoplasias Encefálicas/terapia , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Descubrimiento de Drogas , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Variación Genética , Glioma/diagnóstico , Glioma/etiología , Glioma/terapia , Humanos , Metabolómica/métodos , Terapia Molecular Dirigida , Transducción de Señal
8.
Cell Metab ; 30(3): 525-538.e8, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31303424

RESUMEN

Advances in DNA sequencing technologies have reshaped our understanding of the molecular basis of cancer, providing a precise genomic view of tumors. Complementary biochemical and biophysical perspectives of cancer point toward profound shifts in nutrient uptake and utilization that propel tumor growth and major changes in the structure of the plasma membrane of tumor cells. The molecular mechanisms that bridge these fundamental aspects of tumor biology remain poorly understood. Here, we show that the lysophosphatidylcholine acyltransferase LPCAT1 functionally links specific genetic alterations in cancer with aberrant metabolism and plasma membrane remodeling to drive tumor growth. Growth factor receptor-driven cancers are found to depend on LPCAT1 to shape plasma membrane composition through enhanced saturated phosphatidylcholine content that is, in turn, required for the transduction of oncogenic signals. These results point to a genotype-informed strategy that prioritizes lipid remodeling pathways as therapeutic targets for diverse cancers.


Asunto(s)
1-Acilglicerofosfocolina O-Aciltransferasa/metabolismo , Amplificación de Genes , Neoplasias/genética , Neoplasias/metabolismo , Oncogenes/genética , Fosfolípidos/metabolismo , 1-Acilglicerofosfocolina O-Aciltransferasa/genética , Células A549 , Animales , Supervivencia Celular/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Genotipo , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Células PC-3 , Transducción de Señal/genética , Transfección
9.
Mol Cell Biol ; 36(18): 2403-17, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27354066

RESUMEN

Heat shock factor 1 (HSF1) monitors the structural integrity of the proteome. Phosphorylation at S326 is a hallmark for HSF1 activation, but the identity of the kinase(s) phosphorylating this site has remained elusive. We show here that the dietary agent phenethyl isothiocyanate (PEITC) inhibits heat shock protein 90 (Hsp90), the main negative regulator of HSF1; activates p38 mitogen-activated protein kinase (MAPK); and increases S326 phosphorylation, trimerization, and nuclear translocation of HSF1, and the transcription of a luciferase reporter, as well as the endogenous prototypic HSF1 target Hsp70. In vitro, all members of the p38 MAPK family rapidly and stoichiometrically catalyze the S326 phosphorylation. The use of stable knockdown cell lines and inhibitors indicated that among the p38 MAPKs, p38γ is the principal isoform responsible for the phosphorylation of HSF1 at S326 in cells. A protease-mass spectrometry approach confirmed S326 phosphorylation and unexpectedly revealed that p38 MAPK also catalyzes the phosphorylation of HSF1 at S303/307, previously known repressive posttranslational modifications. Thus, we have identified p38 MAPKs as highly efficient catalysts for the phosphorylation of HSF1. Furthermore, our findings suggest that the magnitude and persistence of activation of p38 MAPK are important determinants of the extent and duration of the heat shock response.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Serina/metabolismo , Factores de Transcripción/genética , Transcripción Genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/química , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Células HeLa , Factores de Transcripción del Choque Térmico , Humanos , Isotiocianatos/farmacología , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Fosforilación , Multimerización de Proteína , Transporte de Proteínas , Factores de Transcripción/química
10.
Nat Commun ; 6: 7066, 2015 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-25967870

RESUMEN

Forebrain neurons have weak intrinsic antioxidant defences compared with astrocytes, but the molecular basis and purpose of this is poorly understood. We show that early in mouse cortical neuronal development in vitro and in vivo, expression of the master-regulator of antioxidant genes, transcription factor NF-E2-related-factor-2 (Nrf2), is repressed by epigenetic inactivation of its promoter. Consequently, in contrast to astrocytes or young neurons, maturing neurons possess negligible Nrf2-dependent antioxidant defences, and exhibit no transcriptional responses to Nrf2 activators, or to ablation of Nrf2's inhibitor Keap1. Neuronal Nrf2 inactivation seems to be required for proper development: in maturing neurons, ectopic Nrf2 expression inhibits neurite outgrowth and aborization, and electrophysiological maturation, including synaptogenesis. These defects arise because Nrf2 activity buffers neuronal redox status, inhibiting maturation processes dependent on redox-sensitive JNK and Wnt pathways. Thus, developmental epigenetic Nrf2 repression weakens neuronal antioxidant defences but is necessary to create an environment that supports neuronal development.


Asunto(s)
Antioxidantes/metabolismo , Corteza Cerebral/citología , Epigénesis Genética/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Factor 2 Relacionado con NF-E2/metabolismo , Neuronas/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Corteza Cerebral/embriología , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Fenómenos Electrofisiológicos , Proteína 1 Asociada A ECH Tipo Kelch , Ratones , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética
11.
Mol Cell Biol ; 34(17): 3305-20, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24958099

RESUMEN

Mice lacking the transcription factor NF-E2 p45-related factor 2 (Nrf2) develop more severe nonalcoholic steatohepatitis (NASH), with cirrhosis, than wild-type (Nrf2(+/+)) mice when fed a high-fat (HF) diet for 24 weeks. Although NASH is usually associated with insulin resistance, HF-fed Nrf2(-/-) mice exhibited better insulin sensitivity than HF-fed Nrf2(+/+) mice. In livers of HF-fed mice, loss of Nrf2 resulted in greater induction of lipogenic genes, lower expression of ß-oxidation genes, greater reduction in AMP-activated protein kinase (AMPK) levels, and diminished acetyl coenzyme A (CoA) carboxylase phosphorylation than in the wild-type livers, which is consistent with greater fatty acid (FA) synthesis in Nrf2(-/-) livers. Moreover, primary Nrf2(-/-) hepatocytes displayed lower glucose and FA oxidation than Nrf2(+/+) hepatocytes, with FA oxidation partially rescued by treatment with AMPK activators. The unfolded protein response (UPR) was perturbed in control regular-chow (RC)-fed Nrf2(-/-) mouse livers, and this was associated with constitutive activation of NF-κB and JNK, along with upregulation of inflammatory genes. The HF diet elicited an antioxidant response in Nrf2(+/+) livers, and as this was compromised in Nrf2(-/-) livers, they suffered oxidative stress. Therefore, Nrf2 protects against NASH by suppressing lipogenesis, supporting mitochondrial function, increasing the threshold for the UPR and inflammation, and enabling adaptation to HF-diet-induced oxidative stress.


Asunto(s)
Cirrosis Hepática Experimental/etiología , Factor 2 Relacionado con NF-E2/deficiencia , Enfermedad del Hígado Graso no Alcohólico/etiología , Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Susceptibilidad a Enfermedades , Estrés del Retículo Endoplásmico , Resistencia a la Insulina , Metabolismo de los Lípidos/genética , Lipogénesis/genética , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática Experimental/genética , Cirrosis Hepática Experimental/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Estrés Oxidativo , Fosforilación , Transducción de Señal , Respuesta de Proteína Desplegada
12.
Mol Cell Biol ; 31(6): 1121-33, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21245377

RESUMEN

Regulation of transcription factor Nrf2 (NF-E2-related factor 2) involves redox-sensitive proteasomal degradation via the E3 ubiquitin ligase Keap1/Cul3. However, Nrf2 is controlled by other mechanisms that have not yet been elucidated. We now show that glycogen synthase kinase 3 (GSK-3) phosphorylates a group of Ser residues in the Neh6 domain of mouse Nrf2 that overlap with an SCF/ß-TrCP destruction motif (DSGIS, residues 334 to 338) and promotes its degradation in a Keap1-independent manner. Nrf2 was stabilized by GSK-3 inhibitors in Keap1-null mouse embryo fibroblasts. Similarly, an Nrf2(ΔETGE) mutant, which cannot be degraded via Keap1, accumulated when GSK-3 activity was blocked. Phosphorylation of a Ser cluster in the Neh6 domain of Nrf2 stimulated its degradation because a mutant Nrf2(ΔETGE 6S/6A) protein, lacking these Ser residues, exhibited a longer half-life than Nrf2(ΔETGE). Moreover, Nrf2(ΔETGE 6S/6A) was insensitive to ß-TrCP regulation and exhibited lower levels of ubiquitination than Nrf2(ΔETGE). GSK-3ß enhanced ubiquitination of Nrf2(ΔETGE) but not that of Nrf2(ΔETGE 6S/6A). The Nrf2(ΔETGE) protein but not Nrf2(ΔETGE 6S/6A) coimmunoprecipitated with ß-TrCP, and this association was enhanced by GSK-3ß. Our results show for the first time that Nrf2 is targeted by GSK-3 for SCF/ß-TrCP-dependent degradation. We propose a "dual degradation" model to describe the regulation of Nrf2 under different pathophysiological conditions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas del Citoesqueleto/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas con Repetición de beta-Transducina/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Glucógeno Sintasa Quinasa 3 beta , Humanos , Proteína 1 Asociada A ECH Tipo Kelch , Ratones , Datos de Secuencia Molecular , Mutación , Factor 2 Relacionado con NF-E2/química , Factor 2 Relacionado con NF-E2/genética , Fosforilación , Estructura Terciaria de Proteína , Serina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
13.
Antioxid Redox Signal ; 13(11): 1713-48, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20446772

RESUMEN

The cap'n'collar (CNC) bZIP transcription factor Nrf2 controls expression of genes for antioxidant enzymes, metal-binding proteins, drug-metabolising enzymes, drug transporters, and molecular chaperones. Many chemicals that protect against carcinogenesis induce Nrf2-target genes. These compounds are all thiol-reactive and stimulate an adaptive response to redox stress in cells. Such agents induce the expression of genes that posses an antioxidant response element (ARE) in their regulatory regions. Under normal homeostatic conditions, Nrf2 activity is restricted through a Keap1-dependent ubiquitylation by Cul3-Rbx1, which targets the CNC-bZIP transcription factor for proteasomal degradation. However, as the substrate adaptor function of Keap1 is redox-sensitive, Nrf2 protein evades ubiquitylation by Cul3-Rbx1 when cells are treated with chemopreventive agents. As a consequence, Nrf2 accumulates in the nucleus where it heterodimerizes with small Maf proteins and transactivates genes regulated through an ARE. In this review, we describe synthetic compounds and phytochemicals from edible plants that induce Nrf2-target genes. We also discuss evidence for the existence of different classes of ARE (a 16-bp 5'-TMAnnRTGABnnnGCR-3' versus an 11-bp 5'-RTGABnnnGCR-3', with or without the embedded activator protein 1-binding site 5'-TGASTCA-3'), species differences in the ARE-gene battery, and the identity of critical Cys residues in Keap1 required for de-repression of Nrf2 by chemopreventive agents.


Asunto(s)
Anticarcinógenos/uso terapéutico , Antioxidantes/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias/prevención & control , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Anticarcinógenos/metabolismo , Antioxidantes/metabolismo , Proteínas Portadoras/metabolismo , Proliferación Celular/efectos de los fármacos , Proteínas Cullin/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína 1 Asociada A ECH Tipo Kelch , Factor 2 Relacionado con NF-E2/genética , Neoplasias/metabolismo , Elementos de Respuesta/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transactivadores/química , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
14.
Free Radic Biol Med ; 48(2): 357-71, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19914374

RESUMEN

Nonalcoholic steatohepatitis (NASH) arises from nonalcoholic fatty liver disease (NAFLD) as a consequence of oxidative stress. Herein we report that the development of NASH is greatly accelerated in mice lacking transcription factor Nrf2 when they are challenged with a methionine- and choline-deficient (MCD) diet. After 14 days of feeding on an MCD diet, livers from Nrf2(-/-) mice showed a substantial increase in macro- and microvesicular steatosis and a massive increase in the number of neutrophil polymorphs, compared to livers from wild-type mice treated similarly. Livers of Nrf2(-/-) mice on the MCD diet suffered more oxidative stress than their wild-type counterparts as assessed by a significant depletion of reduced glutathione that was coupled with increases in oxidized glutathione and malondialdehyde. Furthermore, livers from Nrf2(-/-) mice on the MCD diet suffered heightened inflammation as judged by an approximately 10-fold increase in the amount of nuclear NF-kappaB p65 protein and approximately 5-fold increases in the levels of mRNA for interleukin-1beta, tumor necrosis factor alpha, cyclooxygenase 2, and inducible nitric oxide synthase compared with livers from similarly treated wild-type mice. Thus, impairment of Nrf2 activity may represent a major risk factor for the evolution of NAFLD to NASH.


Asunto(s)
Hígado Graso/metabolismo , Hígado/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Colina/química , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Progresión de la Enfermedad , Hígado Graso/sangre , Hígado Graso/genética , Hígado Graso/patología , Hígado Graso/fisiopatología , Alimentos Formulados , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Hígado/patología , Masculino , Metionina/química , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , FN-kappa B/metabolismo , Infiltración Neutrófila/genética , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Estrés Oxidativo/genética , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA