Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Nat Rev Mol Cell Biol ; 25(1): 46-64, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37710009

RESUMEN

The forkhead box protein O (FOXO, consisting of FOXO1, FOXO3, FOXO4 and FOXO6) transcription factors are the mammalian orthologues of Caenorhabditis elegans DAF-16, which gained notoriety for its capability to double lifespan in the absence of daf-2 (the gene encoding the worm insulin receptor homologue). Since then, research has provided many mechanistic details on FOXO regulation and FOXO activity. Furthermore, conditional knockout experiments have provided a wealth of data as to how FOXOs control development and homeostasis at the organ and organism levels. The lifespan-extending capabilities of DAF-16/FOXO are highly correlated with their ability to induce stress response pathways. Exogenous and endogenous stress, such as cellular redox stress, are considered the main drivers of the functional decline that characterizes ageing. Functional decline often manifests as disease, and decrease in FOXO activity indeed negatively impacts on major age-related diseases such as cancer and diabetes. In this context, the main function of FOXOs is considered to preserve cellular and organismal homeostasis, through regulation of stress response pathways. Paradoxically, the same FOXO-mediated responses can also aid the survival of dysfunctional cells once these eventually emerge. This general property to control stress responses may underlie the complex and less-evident roles of FOXOs in human lifespan as opposed to model organisms such as C. elegans.


Asunto(s)
Caenorhabditis elegans , Transducción de Señal , Animales , Humanos , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Transducción de Señal/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Envejecimiento/genética , Longevidad/genética , Mamíferos/metabolismo
2.
Hum Mol Genet ; 28(1): 96-104, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30239721

RESUMEN

Loss-of-function mutations in glutaminase (GLS), the enzyme converting glutamine into glutamate, and the counteracting enzyme glutamine synthetase (GS) cause disturbed glutamate homeostasis and severe neonatal encephalopathy. We report a de novo Ser482Cys gain-of-function variant in GLS encoding GLS associated with profound developmental delay and infantile cataract. Functional analysis demonstrated that this variant causes hyperactivity and compensatory downregulation of GLS expression combined with upregulation of the counteracting enzyme GS, supporting pathogenicity. Ser482Cys-GLS likely improves the electrostatic environment of the GLS catalytic site, thereby intrinsically inducing hyperactivity. Alignment of +/-12.000 GLS protein sequences from >1000 genera revealed extreme conservation of Ser482 to the same degree as catalytic residues. Together with the hyperactivity, this indicates that Ser482 is evolutionarily preserved to achieve optimal-but submaximal-GLS activity. In line with GLS hyperactivity, increased glutamate and decreased glutamine concentrations were measured in urine and fibroblasts. In the brain (both grey and white matter), glutamate was also extremely high and glutamine was almost undetectable, demonstrated with magnetic resonance spectroscopic imaging at clinical field strength and subsequently supported at ultra-high field strength. Considering the neurotoxicity of glutamate when present in excess, the strikingly high glutamate concentrations measured in the brain provide an explanation for the developmental delay. Cataract, a known consequence of oxidative stress, was evoked in zebrafish expressing the hypermorphic Ser482Cys-GLS and could be alleviated by inhibition of GLS. The capacity to detoxify reactive oxygen species was reduced upon Ser482Cys-GLS expression, providing an explanation for cataract formation. In conclusion, we describe an inborn error of glutamate metabolism caused by a GLS hyperactivity variant, illustrating the importance of balanced GLS activity.


Asunto(s)
Glutaminasa/genética , Glutaminasa/fisiología , Adolescente , Animales , Encéfalo/metabolismo , Catarata/genética , Preescolar , Discapacidades del Desarrollo/genética , Modelos Animales de Enfermedad , Femenino , Fibroblastos , Mutación con Ganancia de Función/genética , Glutamato-Amoníaco Ligasa/genética , Glutamato-Amoníaco Ligasa/fisiología , Ácido Glutámico/genética , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Células HEK293 , Humanos , Masculino , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Pez Cebra
3.
Mol Cell ; 49(4): 730-42, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23333309

RESUMEN

Forkhead box O (FOXO; DAF-16 in worms) transcription factors, which are of vital importance in cell-cycle control, stress resistance, tumor suppression, and organismal lifespan, are largely regulated through nucleo-cytoplasmic shuttling. Insulin signaling keeps FOXO/DAF-16 cytoplasmic, and hence transcriptionally inactive. Conversely, as in loss of insulin signaling, reactive oxygen species (ROS) can activate FOXO/DAF-16 through nuclear accumulation. How ROS regulate the nuclear translocation of FOXO/DAF-16 is largely unknown. Cysteine oxidation can stabilize protein-protein interactions through the formation of disulfide-bridges when cells encounter ROS. Using a proteome-wide screen that identifies ROS-induced mixed disulfide-dependent complexes, we discovered several interaction partners of FOXO4, one of which is the nuclear import receptor transportin-1. We show that disulfide formation with transportin-1 is required for nuclear localization and the activation of FOXO4/DAF-16 induced by ROS, but not by the loss of insulin signaling. This molecular mechanism for nuclear shuttling is conserved in C. elegans and directly connects redox signaling to the longevity protein FOXO/DAF-16.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Factores de Transcripción/metabolismo , beta Carioferinas/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Caenorhabditis elegans/citología , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Cistina/metabolismo , Factores de Transcripción Forkhead , Células HEK293 , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Oxidación-Reducción , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/genética , beta Carioferinas/fisiología
4.
Biochem Soc Trans ; 48(2): 379-397, 2020 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-32311028

RESUMEN

It is well established that both an increase in reactive oxygen species (ROS: i.e. O2•-, H2O2 and OH•), as well as protein aggregation, accompany ageing and proteinopathies such as Parkinson's and Alzheimer's disease. However, it is far from clear whether there is a causal relation between the two. This review describes how protein aggregation can be affected both by redox signalling (downstream of H2O2), as well as by ROS-induced damage, and aims to give an overview of the current knowledge of how redox signalling affects protein aggregation and vice versa. Redox signalling has been shown to play roles in almost every step of protein aggregation and amyloid formation, from aggregation initiation to the rapid oligomerization of large amyloids, which tend to be less toxic than oligomeric prefibrillar aggregates. We explore the hypothesis that age-associated elevated ROS production could be part of a redox signalling-dependent-stress response in an attempt to curb protein aggregation and minimize toxicity.


Asunto(s)
Oxidación-Reducción , Agregado de Proteínas , Transducción de Señal , Envejecimiento , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Humanos , Peróxido de Hidrógeno/química , Ratones , Oxígeno/química , Enfermedad de Parkinson/metabolismo , Unión Proteica , Pliegue de Proteína , Proteostasis , Especies Reactivas de Oxígeno
5.
J Biol Chem ; 288(30): 21729-41, 2013 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-23770673

RESUMEN

FOXO (forkhead box O) transcription factors are tumor suppressors and increase the life spans of model organisms. Cellular stress, in particular oxidative stress caused by an increase in levels of reactive oxygen species (ROS), activates FOXOs through JNK-mediated phosphorylation. Importantly, JNK regulation of FOXO is evolutionarily conserved. Here we identified the pathway that mediates ROS-induced JNK-dependent FOXO regulation. Following increased ROS, RALA is activated by the exchange factor RLF (RalGDS-like factor), which is in complex with JIP1 (C-Jun-amino-terminal-interacting protein 1) and JNK. Active RALA consequently regulates assembly and activation of MLK3, MKK4, and JNK onto the JIP1 scaffold. Furthermore, regulation of FOXO by RALA and JIP1 is conserved in C. elegans, where both ral-1 and jip-1 depletion impairs heat shock-induced nuclear translocation of the FOXO orthologue DAF16.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Unión al GTP ral/metabolismo , Transporte Activo de Núcleo Celular , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Western Blotting , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Activación Enzimática , Factores de Transcripción Forkhead , Células HEK293 , Humanos , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Proteína Quinasa 8 Activada por Mitógenos/genética , Mutación , Células 3T3 NIH , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Proteínas de Unión al GTP ral/genética
6.
Biochem Soc Trans ; 42(4): 971-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25109988

RESUMEN

Until recently, ROS (reactive oxygen species) were often seen as merely damaging agents. However, small, but significant, amounts of hydrogen peroxide (H2O2) are also being produced upon, for instance, NADPH-oxidase activation in response to growth factor signalling and as a by-product of mitochondrial respiration. H2O2 perturbs the local cellular redox state and this results in specific and reversible cysteine oxidation in target proteins, thereby translating the redox state into a signal that ultimately leads to an appropriate cellular response. This phenomenon of signalling through cysteine oxidation is known as redox signalling and has recently been shown to be involved in a wide range of physiological processes. Cysteine residue oxidation can lead to a range of post-translational modifications, one of which is the formation of intermolecular disulfides. In the present mini-review we will give a number of examples of proteins regulated by intermolecular disulfides and discuss a recently developed method to screen for these interactions. The consequences of the regulation of the FOXO4 (forkhead box O4) transcription factor by formation of intermolecular disulfides with both TNPO1 (transportin 1) and p300/CBP [CREB (cAMP-response-element-binding protein)-binding protein] are discussed in more detail.


Asunto(s)
Cisteína/metabolismo , Disulfuros/metabolismo , Factores de Transcripción Forkhead/metabolismo , Animales , Cisteína/química , Factores de Transcripción Forkhead/genética , Humanos , Oxidación-Reducción , Transducción de Señal
7.
Cancer Cell ; 10(2): 113-20, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16904610

RESUMEN

Overexpression of Bcl-xL, loss of p19 ARF, and loss of p53 all accelerate Myc oncogenesis. All three lesions are implicated in suppressing Myc-induced apoptosis, suggesting that this is a common mechanism by which they synergize with Myc. However, using an acutely switchable model of Myc-induced tumorigenesis, we demonstrate that each lesion cooperates with Myc in vivo by a distinct mechanism. While Bcl-xL blocks Myc-induced apoptosis, inactivation of p19 ARF enhances it. However, this increase in apoptosis is matched by increased Myc-induced proliferation. p53 inactivation shares features of both lesions, partially suppressing apoptosis while augmenting proliferation. Bcl-xL and p19 ARF loss together synergize to further accelerate Myc oncogenesis. Thus, differing lesions cooperate oncogenically with Myc by discrete mechanisms that can themselves synergize with each other.


Asunto(s)
Apoptosis , Transformación Celular Neoplásica , Genes myc/fisiología , Proteína p14ARF Supresora de Tumor/fisiología , Proteína bcl-X/fisiología , Animales , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Secretoras de Insulina/patología , Ratones , Ratones Transgénicos , Proteína p53 Supresora de Tumor/fisiología
8.
Free Radic Biol Med ; 223: 369-383, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39059513

RESUMEN

Basic Helix-Loop-Helix (bHLH) transcription factors TFEB/TFE3 and HLH-30 are key regulators of autophagy induction and lysosomal biogenesis in mammals and C. elegans, respectively. While much is known about the regulation of TFEB/TFE3, how HLH-30 subcellular dynamics and transactivation are modulated are yet poorly understood. Thus, elucidating the regulation of C. elegans HLH-30 will provide evolutionary insight into the mechanisms governing the function of bHLH transcription factor family. We report here that HLH-30 is retained in the cytoplasm mainly through its conserved Ser201 residue and that HLH-30 physically interacts with the 14-3-3 protein FTT-2 in this location. The FoxO transcription factor DAF-16 is not required for HLH-30 nuclear translocation upon stress, despite that both proteins partner to form a complex that coordinately regulates several organismal responses. Similar as described for DAF-16, the importin IMB-2 assists HLH-30 nuclear translocation, but constitutive HLH-30 nuclear localization is not sufficient to trigger its distinctive transcriptional response. Furthermore, we identify FTT-2 as the target of diethyl maleate (DEM), a GSH depletor that causes a transient nuclear translocation of HLH-30. Together, our work demonstrates that the regulation of TFEB/TFE3 and HLH-30 family members is evolutionarily conserved and that, in addition to a direct redox regulation through its conserved single cysteine residue, HLH-30 can also be indirectly regulated by a redox-dependent mechanism, probably through FTT-2 oxidation.

9.
Nat Commun ; 15(1): 2725, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38548751

RESUMEN

Reactive Oxygen Species (ROS) derived from mitochondrial respiration are frequently cited as a major source of chromosomal DNA mutations that contribute to cancer development and aging. However, experimental evidence showing that ROS released by mitochondria can directly damage nuclear DNA is largely lacking. In this study, we investigated the effects of H2O2 released by mitochondria or produced at the nucleosomes using a titratable chemogenetic approach. This enabled us to precisely investigate to what extent DNA damage occurs downstream of near- and supraphysiological amounts of localized H2O2. Nuclear H2O2 gives rise to DNA damage and mutations and a subsequent p53 dependent cell cycle arrest. Mitochondrial H2O2 release shows none of these effects, even at levels that are orders of magnitude higher than what mitochondria normally produce. We conclude that H2O2 released from mitochondria is unlikely to directly damage nuclear genomic DNA, limiting its contribution to oncogenic transformation and aging.


Asunto(s)
Peróxido de Hidrógeno , Mitocondrias , Especies Reactivas de Oxígeno/metabolismo , Peróxido de Hidrógeno/metabolismo , Mitocondrias/metabolismo , ADN/metabolismo , Daño del ADN , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo
10.
Free Radic Biol Med ; 206: 134-142, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37392950

RESUMEN

Reactive Oxygen Species (ROS) in the form of H2O2 can act both as physiological signaling molecules as well as damaging agents, depending on their concentration and localization. The downstream biological effects of H2O2 were often studied making use of exogenously added H2O2, generally as a bolus and at supraphysiological levels. But this does not mimic the continuous, low levels of intracellular H2O2 production by for instance mitochondrial respiration. The enzyme d-Amino Acid Oxidase (DAAO) catalyzes H2O2 formation using d-amino acids, which are absent from culture media, as a substrate. Ectopic expression of DAAO has recently been used in several studies to produce inducible and titratable intracellular H2O2. However, a method to directly quantify the amount of H2O2 produced by DAAO has been lacking, making it difficult to assess whether observed phenotypes are the result of physiological or artificially high levels of H2O2. Here we describe a simple assay to directly quantify DAAO activity by measuring the oxygen consumed during H2O2 production. The oxygen consumption rate (OCR) of DAAO can directly be compared to the basal mitochondrial respiration in the same assay, to estimate whether the ensuing level of H2O2 production is within the range of physiological mitochondrial ROS production. In the tested monoclonal RPE1-hTERT cells, addition of 5 mM d-Ala to the culture media amounts to a DAAO-dependent OCR that surpasses ∼5% of the OCR that stems from basal mitochondrial respiration and hence produces supra-physiological levels of H2O2. We show that the assay can also be used to select clones that express differentially localized DAAO with the same absolute level of H2O2 production to be able to discriminate the effects of H2O2 production at different subcellular locations from differences in total oxidative burden. This method therefore greatly improves the interpretation and applicability of DAAO-based models, thereby moving the redox biology field forward.


Asunto(s)
Aminoácidos , Peróxido de Hidrógeno , Humanos , Peróxido de Hidrógeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Aminoácidos/metabolismo , Consumo de Oxígeno , Oxígeno
12.
Nat Chem Biol ; 5(9): 664-72, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19648934

RESUMEN

Cellular damage invoked by reactive oxygen species plays a key role in the pathobiology of cancer and aging. Forkhead box class O (FoxO) transcription factors are involved in various cellular processes including cell cycle regulation, apoptosis and resistance to reactive oxygen species, and studies in animal models have shown that these transcription factors are of vital importance in tumor suppression, stem cell maintenance and lifespan extension. Here we report that the activity of FoxO in human cells is directly regulated by the cellular redox state through a unique mechanism in signal transduction. We show that reactive oxygen species induce the formation of cysteine-thiol disulfide-dependent complexes of FoxO and the p300/CBP acetyltransferase, and that modulation of FoxO biological activity by p300/CBP-mediated acetylation is fully dependent on the formation of this redox-dependent complex. These findings directly link cellular redox status to the activity of the longevity protein FoxO.


Asunto(s)
Cisteína/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Animales , Proteínas de Ciclo Celular , Línea Celular , Supervivencia Celular , Cisteína/genética , Factores de Transcripción Forkhead , Humanos , Lisina/genética , Lisina/metabolismo , Ratones , Mutación , Oxidación-Reducción , Peróxidos/farmacología , Transducción de Señal , Tiorredoxinas/farmacología , Factores de Transcripción/genética , Factores de Transcripción p300-CBP/genética
13.
STAR Protoc ; 2(1): 100273, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33490987

RESUMEN

The relative positioning of organelles underlies fundamental cellular processes, including signaling, polarization, and cellular growth. Here, we describe the usage of a light-dependent heterodimerization system, LOVpep-ePDZ, to alter organelle positioning locally and reversibly in order to study the functional consequences of organelle positioning. The protocol gives details on how to accomplish expression of fusion proteins encoding this system, describes the imaging parameters to achieve subcellular activation in C. elegans, and may be adapted for use in other model systems. For complete details on the use and execution of this protocol, please refer to De Henau et al. (2020).


Asunto(s)
Animales Modificados Genéticamente , Caenorhabditis elegans , Optogenética , Orgánulos , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Dimerización , Orgánulos/genética , Orgánulos/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética
14.
Free Radic Biol Med ; 172: 298-311, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34144191

RESUMEN

Stabilization and activation of the p53 tumor suppressor are triggered in response to various cellular stresses, including DNA damaging agents and elevated Reactive Oxygen Species (ROS) like H2O2. When cells are exposed to exogenously added H2O2, ATR/CHK1 and ATM/CHK2 dependent DNA damage signaling is switched on, suggesting that H2O2 induces both single and double strand breaks. These collective observations have resulted in the widely accepted model that oxidizing conditions lead to DNA damage that subsequently mediates a p53-dependent response like cell cycle arrest and apoptosis. However, H2O2 also induces signaling through stress-activated kinases (SAPK, e.g., JNK and p38 MAPK) that can activate p53. Here we dissect to what extent these pathways contribute to functional activation of p53 in response to oxidizing conditions. Collectively, our data suggest that p53 can be activated both by SAPK signaling and the DDR independently of each other, and which of these pathways is activated depends on the type of oxidant used. This implies that it could in principle be possible to modulate oxidative signaling to stimulate p53 without inducing collateral DNA damage, thereby limiting mutation accumulation in both healthy and tumor tissues.


Asunto(s)
Proteínas de Ciclo Celular , Proteína p53 Supresora de Tumor , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Humanos , Peróxido de Hidrógeno , Oxidantes/farmacología , Fosforilación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
15.
Antioxidants (Basel) ; 10(10)2021 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-34679713

RESUMEN

Reversible cysteine oxidation plays an essential role in redox signaling by reversibly altering protein structure and function. Cysteine oxidation may lead to intra- and intermolecular disulfide formation, and the latter can drastically stabilize protein-protein interactions in a more oxidizing milieu. The activity of the tumor suppressor p53 is regulated at multiple levels, including various post-translational modification (PTM) and protein-protein interactions. In the past few decades, p53 has been shown to be a redox-sensitive protein, and undergoes reversible cysteine oxidation both in vitro and in vivo. It is not clear, however, whether p53 also forms intermolecular disulfides with interacting proteins and whether these redox-dependent interactions contribute to the regulation of p53. In the present study, by combining (co-)immunoprecipitation, quantitative mass spectrometry and Western blot we found that p53 forms disulfide-dependent interactions with several proteins under oxidizing conditions. Cysteine 277 is required for most of the disulfide-dependent interactions of p53, including those with 14-3-3θ and 53BP1. These interaction partners may play a role in fine-tuning p53 activity under oxidizing conditions.

16.
Antioxidants (Basel) ; 10(4)2021 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-33923941

RESUMEN

Redox signaling is controlled by the reversible oxidation of cysteine thiols, a post-translational modification triggered by H2O2 acting as a second messenger. However, H2O2 actually reacts poorly with most cysteine thiols and it is not clear how H2O2 discriminates between cysteines to trigger appropriate signaling cascades in the presence of dedicated H2O2 scavengers like peroxiredoxins (PRDXs). It was recently suggested that peroxiredoxins act as peroxidases and facilitate H2O2-dependent oxidation of redox-regulated proteins via disulfide exchange reactions. It is unknown how the peroxiredoxin-based relay model achieves the selective substrate targeting required for adequate cellular signaling. Using a systematic mass-spectrometry-based approach to identify cysteine-dependent interactors of the five human 2-Cys peroxiredoxins, we show that all five human 2-Cys peroxiredoxins can form disulfide-dependent heterodimers with a large set of proteins. Each isoform displays a preference for a subset of disulfide-dependent binding partners, and we explore isoform-specific properties that might underlie this preference. We provide evidence that peroxiredoxin-based redox relays can proceed via two distinct molecular mechanisms. Altogether, our results support the theory that peroxiredoxins could play a role in providing not only reactivity but also selectivity in the transduction of peroxide signals to generate complex cellular signaling responses.

17.
Cell Rep ; 34(4): 108675, 2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33503422

RESUMEN

DNA replication is challenged by numerous exogenous and endogenous factors that can interfere with the progression of replication forks. Substantial accumulation of single-stranded DNA during DNA replication activates the DNA replication stress checkpoint response that slows progression from S/G2 to M phase to protect genomic integrity. Whether and how mild replication stress restricts proliferation remains controversial. Here, we identify a cell cycle exit mechanism that prevents S/G2 phase arrested cells from undergoing mitosis after exposure to mild replication stress through premature activation of the anaphase promoting complex/cyclosome (APC/CCDH1). We find that replication stress causes a gradual decrease of the levels of the APC/CCDH1 inhibitor EMI1/FBXO5 through Forkhead box O (FOXO)-mediated inhibition of its transcription factor E2F1. By doing so, FOXOs limit the time during which the replication stress checkpoint is reversible and thereby play an important role in maintaining genomic stability.


Asunto(s)
Ciclo Celular/fisiología , Daño del ADN/genética , Replicación del ADN/genética , Inestabilidad Genómica/genética , Proliferación Celular , Humanos
18.
Antioxid Redox Signal ; 33(12): 839-859, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-32151151

RESUMEN

Significance: The p53 tumor suppressor has been dubbed the "guardian of genome" because of its various roles in the response to DNA damage such as DNA damage repair, cell cycle arrest, senescence, and apoptosis, all of which are in place to prevent mutations from being passed on down the lineage. Recent Advances: Reactive oxygen species (ROS), for instance hydrogen peroxide derived from mitochondrial respiration, have long been regarded mainly as a major source of cellular damage to DNA and other macromolecules. Critical Issues: More recently, ROS have been shown to also play important physiological roles as second messengers in so-called redox signaling. It is, therefore, not clear whether the observed activation of p53 by ROS is mediated through the DNA damage response, redox signaling, or both. In this review, we will discuss the similarities and differences between p53 activation in response to DNA damage and redox signaling in terms of upstream signaling and downstream transcriptional program activation. Future Directions: Understanding whether and how DNA damage and redox signaling-dependent p53 activation can be dissected could be useful to develop anti-cancer therapeutic p53-reactivation strategies that do not depend on the induction of DNA damage and the resulting additional mutational load.


Asunto(s)
Daño del ADN , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Respiración de la Célula , Cisteína/metabolismo , Reparación del ADN , Humanos , Peróxido de Hidrógeno/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo
19.
Mol Biol Cell ; 31(14): 1486-1497, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32374641

RESUMEN

Actomyosin-based contractility in smooth muscle and nonmuscle cells is regulated by signaling through the small GTPase Rho and by calcium-activated pathways. We use the myoepithelial cells of the Caenorhabditis elegans spermatheca to study the mechanisms of coordinated myosin activation in vivo. Here, we show that redox signaling modulates RHO-1/Rho activity in this contractile tissue. Exogenously added as well as endogenously generated hydrogen peroxide decreases spermathecal contractility by inhibition of RHO-1, which depends on a conserved cysteine in its nucleotide binding site (C20). Further, we identify an endogenous gradient of H2O2 across the spermathecal tissue, which depends on the activity of cytosolic superoxide dismutase, SOD-1. Collectively, we show that SOD-1-mediated H2O2 production regulates the redox environment and fine tunes Rho activity across the spermatheca through oxidation of RHO-1 C20.


Asunto(s)
Células Epiteliales/metabolismo , Contracción Muscular/fisiología , Proteínas de Unión al GTP rho/metabolismo , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Calcio/metabolismo , Peróxido de Hidrógeno/metabolismo , Células Musculares/metabolismo , Músculo Liso/metabolismo , Músculo Liso/fisiología , Oxidación-Reducción , Transducción de Señal , Superóxido Dismutasa/metabolismo , Quinasas Asociadas a rho/metabolismo
20.
Dev Cell ; 53(3): 263-271.e6, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32275886

RESUMEN

Symmetry breaking is an essential step in cell differentiation and early embryonic development. However, the molecular cues that trigger symmetry breaking remain largely unknown. Here, we show that mitochondrial H2O2 acts as a symmetry-breaking cue in the C. elegans zygote. We find that symmetry breaking is marked by a local H2O2 increase and coincides with a relocation of mitochondria to the cell cortex. Lowering endogenous H2O2 levels delays the onset of symmetry breaking, while artificially targeting mitochondria to the cellular cortex using a light-induced heterodimerization technique is sufficient to initiate symmetry breaking in a H2O2-dependent manner. In wild-type development, both sperm and maternal mitochondria contribute to symmetry breaking. Our findings reveal that mitochondrial H2O2-signaling promotes the onset of polarization, a fundamental process in development and cell differentiation, and this is achieved by both mitochondrial redistribution and differential H2O2-production.


Asunto(s)
Tipificación del Cuerpo/efectos de los fármacos , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/embriología , Embrión no Mamífero/citología , Peróxido de Hidrógeno/farmacología , Mitocondrias/metabolismo , Cigoto/citología , Animales , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/fisiología , Proteínas de Caenorhabditis elegans/genética , Polaridad Celular , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Oxidantes/farmacología , Cigoto/efectos de los fármacos , Cigoto/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA