Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 42(1): 111997, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36656716

RESUMEN

Nitric oxide (NO) production in the tumor microenvironment is a common element in cancer. S-nitrosylation, the post-translational modification of cysteines by NO, is emerging as a key transduction mechanism sustaining tumorigenesis. However, most oncoproteins that are regulated by S-nitrosylation are still unknown. Here we show that S-nitrosoglutathione reductase (GSNOR), the enzyme that deactivates S-nitrosylation, is hypo-expressed in several human malignancies. Using multiple tumor models, we demonstrate that GSNOR deficiency induces S-nitrosylation of focal adhesion kinase 1 (FAK1) at C658. This event enhances FAK1 autophosphorylation and sustains tumorigenicity by providing cancer cells with the ability to survive in suspension (evade anoikis). In line with these results, GSNOR-deficient tumor models are highly susceptible to treatment with FAK1 inhibitors. Altogether, our findings advance our understanding of the oncogenic role of S-nitrosylation, define GSNOR as a tumor suppressor, and point to GSNOR hypo-expression as a therapeutically exploitable vulnerability in cancer.


Asunto(s)
Alcohol Deshidrogenasa , Quinasa 1 de Adhesión Focal , Neoplasias , Humanos , Aldehído Oxidorreductasas/metabolismo , Quinasa 1 de Adhesión Focal/genética , Neoplasias/genética , Óxido Nítrico/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Microambiente Tumoral , Alcohol Deshidrogenasa/metabolismo
2.
Cell Death Dis ; 13(4): 402, 2022 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-35461310

RESUMEN

Tendons are vital collagen-dense specialized connective tissues transducing the force from skeletal muscle to the bone, thus enabling movement of the human body. Tendon cells adjust matrix turnover in response to physiological tissue loading and pathological overloading (tendinopathy). Nevertheless, the regulation of tendon matrix quality control is still poorly understood and the pathogenesis of tendinopathy is presently unsolved. Autophagy, the major mechanism of degradation and recycling of cellular components, plays a fundamental role in the homeostasis of several tissues. Here, we investigate the contribution of autophagy to human tendons' physiology, and we provide in vivo evidence that it is an active process in human tendon tissue. We show that selective autophagy of the endoplasmic reticulum (ER-phagy), regulates the secretion of type I procollagen (PC1), the major component of tendon extracellular matrix. Pharmacological activation of autophagy by inhibition of mTOR pathway alters the ultrastructural morphology of three-dimensional tissue-engineered tendons, shifting collagen fibrils size distribution. Moreover, autophagy induction negatively affects the biomechanical properties of the tissue-engineered tendons, causing a reduction in mechanical strength under tensile force. Overall, our results provide the first evidence that autophagy regulates tendon homeostasis by controlling PC1 quality control, thus potentially playing a role in the development of injured tendons.


Asunto(s)
Autofagia , Tendinopatía , Tendones , Autofagia/fisiología , Colágeno/metabolismo , Colágeno/fisiología , Homeostasis , Humanos , Tendinopatía/metabolismo , Tendinopatía/patología , Tendones/metabolismo , Tendones/patología
3.
Nitric Oxide ; 122-123: 1-5, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35182743

RESUMEN

S-nitrosylation of proteins is a nitric oxide (NO)-based post-translational modification of cysteine residues. By removing the NO moiety from S-nitrosothiol adducts, denitrosylases restore sulfhydryl protein pool and act as downstream tuners of S-nitrosylation signaling. Alterations in the S-nitrosylation/denitrosylation dynamics are implicated in many pathological states, including cancer ontogenesis and progression, skeletal muscle myogenesis and function. Here, we aim to provide and link different lines of evidence, and elaborate on the possible role of S-nitrosylation/denitrosylation signaling in rhabdomyosarcoma, one of the most common pediatric mesenchymal malignancy.


Asunto(s)
Rabdomiosarcoma , S-Nitrosotioles , Niño , Humanos , Desarrollo de Músculos , Óxido Nítrico/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas/metabolismo , S-Nitrosotioles/metabolismo
4.
Nature ; 592(7856): 799-803, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33854232

RESUMEN

Mammalian development, adult tissue homeostasis and the avoidance of severe diseases including cancer require a properly orchestrated cell cycle, as well as error-free genome maintenance. The key cell-fate decision to replicate the genome is controlled by two major signalling pathways that act in parallel-the MYC pathway and the cyclin D-cyclin-dependent kinase (CDK)-retinoblastoma protein (RB) pathway1,2. Both MYC and the cyclin D-CDK-RB axis are commonly deregulated in cancer, and this is associated with increased genomic instability. The autophagic tumour-suppressor protein AMBRA1 has been linked to the control of cell proliferation, but the underlying molecular mechanisms remain poorly understood. Here we show that AMBRA1 is an upstream master regulator of the transition from G1 to S phase and thereby prevents replication stress. Using a combination of cell and molecular approaches and in vivo models, we reveal that AMBRA1 regulates the abundance of D-type cyclins by mediating their degradation. Furthermore, by controlling the transition from G1 to S phase, AMBRA1 helps to maintain genomic integrity during DNA replication, which counteracts developmental abnormalities and tumour growth. Finally, we identify the CHK1 kinase as a potential therapeutic target in AMBRA1-deficient tumours. These results advance our understanding of the control of replication-phase entry and genomic integrity, and identify the AMBRA1-cyclin D pathway as a crucial cell-cycle-regulatory mechanism that is deeply interconnected with genomic stability in embryonic development and tumorigenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ciclina D/metabolismo , Inestabilidad Genómica , Fase S , Animales , Línea Celular , Proliferación Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Replicación del ADN , Regulación del Desarrollo de la Expresión Génica , Genes Supresores de Tumor , Humanos , Ratones , Ratones Noqueados , Mutaciones Letales Sintéticas
5.
J Proteome Res ; 19(10): 4137-4144, 2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-32822197

RESUMEN

Tendon is a highly organized, dense connective tissue that has been demonstrated to have very little turnover. In spite of the low turnover, tendon can grow in response to loading, which may take place primarily at the periphery. Tendon injuries and recurrence of injuries are common in both humans and animals in sports. It is unclear why some areas of the tendon are more susceptible to such injuries and whether this is due to intrinsic regional differences in extracellular matrix (ECM) production or tissue turnover. This study aimed to compare populations of tenocytes derived from the tendon core and periphery. Tenocytes were isolated from equine superficial digital flexor tendons (SDFTs), and the proliferation capacity was determined. ECM production was characterized by immuno- and histological staining and by liquid chromatography-mass spectrometry-based proteomics. Core and periphery SDFT cultures exhibited comparable proliferation rates and had very similar proteome profiles, but showed biological variation in collagen type I deposition. In conclusion, the intrinsic properties of tenocytes from different regions of the tendon are very similar, and other factors in the tissue may contribute to how specific areas respond to loading or injury.


Asunto(s)
Traumatismos de los Tendones , Tenocitos , Animales , Matriz Extracelular , Caballos , Humanos , Proteómica , Tendones
6.
Antioxid Redox Signal ; 32(12): 884-905, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-31931592

RESUMEN

Significance: Cysteines have an essential role in redox signaling, transforming an oxidant signal into a biological response. Among reversible cysteine post-translational modifications, S-nitrosylation acts as a redox-switch in several pathophysiological states, such as ischemia/reperfusion, synaptic transmission, cancer, and muscular dysfunctions. Recent Advances: Growing pieces of in vitro and in vivo evidence argue for S-nitrosylation being deeply involved in development and aging, and playing a role in the onset of different pathological states. New findings suggest it being an enzymatically regulated cellular process, with deep impact on mitochondrial structure and function, and in cellular metabolism. In light of this, the recent discovery of the denitrosylase S-nitrosoCoA (coenzyme A) reductase takes on even greater importance and opens new perspectives on S-nitrosylation as a general mechanism of cellular homeostasis. Critical Issues: Based on these recent findings, we aim at summarizing and elaborating on the established and emerging crucial roles of S-nitrosylation in mitochondrial metabolism and mitophagy, and provide an overview of the pathophysiological effects induced by its deregulation. Future Directions: The identification of new S-nitrosylation targets, and the comprehension of the mechanisms through which S-nitrosylation modulates specific classes of proteins, that is, those impinging on diverse mitochondrial functions, may help to better understand the pathophysiology of aging, and propose lines of intervention to slow down or extend the onset of aging-related diseases.


Asunto(s)
Envejecimiento/metabolismo , Mitocondrias/metabolismo , Enfermedades Musculares/metabolismo , Neoplasias/metabolismo , Óxido Nítrico/metabolismo , Daño por Reperfusión/metabolismo , Envejecimiento/patología , Animales , Humanos , Enfermedades Musculares/patología , Neoplasias/patología , Daño por Reperfusión/patología , Transducción de Señal , Transmisión Sináptica
7.
Cell Death Dis ; 10(5): 354, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31043586

RESUMEN

Neuronal nitric oxide synthase (nNOS) plays a crucial role in the maintenance of correct skeletal muscle function due, at least in part, to S-nitrosylation of specific protein targets. Similarly, we recently provided evidence for a muscular phenotype in mice lacking the denitrosylase S-nitrosoglutathione reductase (GSNOR). Here, we demonstrate that nNOS and GSNOR are concomitantly expressed during differentiation of C2C12. They colocalizes at the sarcolemma and co-immunoprecipitate in cells and in myofibers. We also provide evidence that GSNOR expression decreases in mouse models of muscular dystrophies and of muscle atrophy and wasting, i.e., aging and amyotrophic lateral sclerosis, suggesting a more general regulatory role of GSNOR in skeletal muscle homeostasis.


Asunto(s)
Envejecimiento/genética , Alcohol Deshidrogenasa/genética , Homeostasis/genética , Desarrollo de Músculos/genética , Distrofias Musculares/genética , Óxido Nítrico Sintasa de Tipo I/genética , Envejecimiento/metabolismo , Alcohol Deshidrogenasa/antagonistas & inhibidores , Alcohol Deshidrogenasa/deficiencia , Animales , Diferenciación Celular , Línea Celular Transformada , Modelos Animales de Enfermedad , Proteínas Asociadas a la Distrofina/genética , Proteínas Asociadas a la Distrofina/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Distrofias Musculares/metabolismo , Distrofias Musculares/patología , Mioblastos/citología , Mioblastos/enzimología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Sarcolema/enzimología , Transducción de Señal , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo
8.
Proc Natl Acad Sci U S A ; 115(15): E3388-E3397, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29581312

RESUMEN

S-nitrosylation, a prototypic redox-based posttranslational modification, is frequently dysregulated in disease. S-nitrosoglutathione reductase (GSNOR) regulates protein S-nitrosylation by functioning as a protein denitrosylase. Deficiency of GSNOR results in tumorigenesis and disrupts cellular homeostasis broadly, including metabolic, cardiovascular, and immune function. Here, we demonstrate that GSNOR expression decreases in primary cells undergoing senescence, as well as in mice and humans during their life span. In stark contrast, exceptionally long-lived individuals maintain GSNOR levels. We also show that GSNOR deficiency promotes mitochondrial nitrosative stress, including excessive S-nitrosylation of Drp1 and Parkin, thereby impairing mitochondrial dynamics and mitophagy. Our findings implicate GSNOR in mammalian longevity, suggest a molecular link between protein S-nitrosylation and mitochondria quality control in aging, and provide a redox-based perspective on aging with direct therapeutic implications.


Asunto(s)
Envejecimiento/metabolismo , Mamíferos/metabolismo , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Mitofagia , Envejecimiento/genética , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Animales , Senescencia Celular , Humanos , Mamíferos/genética , Ratones , Ratones Endogámicos C57BL , Mitocondrias/genética , Óxido Nítrico/metabolismo , Estrés Nitrosativo , Procesamiento Proteico-Postraduccional , S-Nitrosotioles/metabolismo
9.
Cancer Res ; 76(14): 4170-82, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27216192

RESUMEN

S-nitrosoglutathione reductase (GSNOR) represents the best-documented denitrosylase implicated in regulating the levels of proteins posttranslationally modified by nitric oxide on cysteine residues by S-nitrosylation. GSNOR controls a diverse array of physiologic functions, including cellular growth and differentiation, inflammation, and metabolism. Chromosomal deletion of GSNOR results in pathologic protein S-nitrosylation that is implicated in human hepatocellular carcinoma (HCC). Here we identify a metabolic hallmark of aberrant S-nitrosylation in HCC and exploit it for therapeutic gain. We find that hepatocyte GSNOR deficiency is characterized by mitochondrial alteration and by marked increases in succinate dehydrogenase (SDH) levels and activity. We find that this depends on the selective S-nitrosylation of Cys(501) in the mitochondrial chaperone TRAP1, which mediates its degradation. As a result, GSNOR-deficient cells and tumors are highly sensitive to SDH inhibition, namely to α-tocopheryl succinate, an SDH-targeting molecule that induced RIP1/PARP1-mediated necroptosis and inhibited tumor growth. Our work provides a specific molecular signature of aberrant S-nitrosylation in HCC, a novel molecular target in SDH, and a first-in-class therapy to treat the disease. Cancer Res; 76(14); 4170-82. ©2016 AACR.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Proteínas HSP90 de Choque Térmico/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Mitocondrias/metabolismo , Succinato Deshidrogenasa/antagonistas & inhibidores , Aldehído Oxidorreductasas/fisiología , Animales , Carcinoma Hepatocelular/metabolismo , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo
10.
FEBS J ; 283(21): 3857-3869, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27083138

RESUMEN

Autophagy is the main catabolic cellular process through which cells adapt their needs (e.g., growth and proliferation) to environmental availability of nutrients (e.g., amino acid and glucose) and growth factors. The rapid activation of the autophagy response essentially depends on protein post-translational modifications (PTMs), which act as molecular switches triggering signaling cascades. Deregulation of autophagy contributes to pathological conditions, such as cancer and neurodegeneration. Therefore, understanding how PTMs affect the occurrence of autophagy is of the highest importance for clinical applications. Besides phosphorylation and ubiquitylation, which represent the best known examples of PTMs, redox-based modifications are also emerging as contributing to the regulation of intracellular signaling. Of note, S-nitrosylation of cysteine residues is a redox PTM and is the principal mechanism of nitric oxide-based signaling. Results emerging in recent years suggest that NO has a role in modulating autophagy. However, the function of S-nitrosylation in autophagy regulation remains still unveiled. By this review, we describe the upstream events regulating autophagy activation focusing on recently published evidence implying a S-nitrosylation-dependent regulation.


Asunto(s)
Autofagia/fisiología , Cisteína/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal/fisiología , Aminoácidos/metabolismo , Glucosa/metabolismo , Humanos , Modelos Biológicos , Oxidación-Reducción , Procesamiento Proteico-Postraduccional
11.
Mol Neurobiol ; 53(6): 4247-4260, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26223801

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder characterized by dopaminergic neuron loss, with an etiopathogenesis involving both genetic and environmental factors. The occupational/residential exposure to the electromagnetic fields has been recently associated with an increased risk of neurodegenerative diseases; it has been thus proposed that the extremely low frequency magnetic field (ELF-MF) may contribute to neurodegenerative etiopathogenesis, as its interaction with biological systems directly impairs redox homeostasis in specific areas of the brain. The molecular mechanisms elicited by ELF-MF, and their potential involvement in PD onset, still remain unclear. To this end, we set up a generator of ELF-MF able to stably and homogeneously reproduce environmental prolonged exposure to ELF-MF (50 Hz, 1 mT). Results obtained indicate that ELF-MF exposure alters cell response of SH-SY5Y cells to MPP(+). We demonstrate that ELF-MF does not affect per se survival, shape, and morphology of both proliferating and differentiated SH-SY5Y cells but significantly impairs redox homeostasis and thiol content, triggering an increase in protein carbonylation. As a result, toxicity of MPP(+), even at low doses, is highly enhanced in ELF-MF-exposed cells due to a significant increase in ROS levels, potentiation of oxidative damage, and induction of a caspase-dependent apoptosis. Pre-incubation with the thiol antioxidants N-acetyl-L-cysteine and GSH ethyl-ester significantly reduces the extent of oxidative damage and protects cells from death induced by the combined treatment ELF-MF/MPP(+). Taken overall, our results demonstrate the redox-based molecular interaction between ELF-MF and PD neurotoxins in vitro, and open a new scenario for defining the synergy of environmental factors in PD onset.


Asunto(s)
1-Metil-4-fenilpiridinio/toxicidad , Campos Magnéticos , Enfermedad de Parkinson/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Homeostasis/efectos de los fármacos , Humanos , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Carbonilación Proteica/efectos de los fármacos , Compuestos de Sulfhidrilo/metabolismo
12.
Mediators Inflamm ; 2015: 536238, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26491229

RESUMEN

Oxidative and nitrosative stresses have been reported as detrimental phenomena concurring to the onset of several neurodegenerative diseases. Here we reported that the ectopic modulation of the denitrosylating enzyme S-nitrosoglutathione reductase (GSNOR) differently impinges on the phenotype of two SH-SY5Y-based in vitro models of neurodegeneration, namely, Parkinson's disease (PD) and familial amyotrophic lateral sclerosis (fALS). In particular, we provide evidence that GSNOR-knocking down protects SH-SY5Y against PD toxins, while, by contrast, its upregulation is required for G93A-SOD1 expressing cells resistance to NO-releasing drugs. Although completely opposite, both conditions are characterized by Nrf2 localization in the nuclear compartment: in the first case induced by GSNOR silencing, while in the second one underlying the antinitrosative response. Overall, our results demonstrate that GSNOR expression has different effect on neuronal viability in dependence on the stimulus applied and suggest that GSNOR could be a responsive gene downstream of Nrf2 activation.


Asunto(s)
Aldehído Oxidorreductasas/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Enfermedad de Parkinson/metabolismo , Animales , Encéfalo/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Femenino , Silenciador del Gen , Humanos , Masculino , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Mitocondrias/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Estrés Oxidativo , Fenotipo , ARN Interferente Pequeño/metabolismo , Médula Espinal/metabolismo
13.
Antioxid Redox Signal ; 21(4): 570-87, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24684653

RESUMEN

AIMS: Nitric oxide (NO) production is implicated in muscle contraction, growth and atrophy, and in the onset of neuropathy. However, many aspects of the mechanism of action of NO are not yet clarified, mainly regarding its role in muscle wasting. Notably, whether NO production-associated neuromuscular atrophy depends on tyrosine nitration or S-nitrosothiols (SNOs) formation is still a matter of debate. Here, we aim at assessing this issue by characterizing the neuromuscular phenotype of S-nitrosoglutathione reductase-null (GSNOR-KO) mice that maintain the capability to produce NO, but are unable to reduce SNOs. RESULTS: We demonstrate that, without any sign of protein nitration, young GSNOR-KO mice show neuromuscular atrophy due to loss of muscle mass, reduced fiber size, and neuropathic behavior. In particular, GSNOR-KO mice show a significant decrease in nerve axon number, with the myelin sheath appearing disorganized and reduced, leading to a dramatic development of a neuropathic phenotype. Mitochondria appear fragmented and depolarized in GSNOR-KO myofibers and myotubes, conditions that are reverted by N-acetylcysteine treatment. Nevertheless, although atrogene transcription is induced, and bulk autophagy activated, no removal of damaged mitochondria is observed. These events, alongside basal increase of apoptotic markers, contribute to persistence of a neuropathic and myopathic state. INNOVATION: Our study provides the first evidence that GSNOR deficiency, which affects exclusively SNOs reduction without altering nitrotyrosine levels, results in a clinically relevant neuromuscular phenotype. CONCLUSION: These findings provide novel insights into the involvement of GSNOR and S-nitrosylation in neuromuscular atrophy and neuropathic pain that are associated with pathological states; for example, diabetes and cancer.


Asunto(s)
Glutatión Reductasa/deficiencia , Enfermedades Neuromusculares/genética , Enfermedades Neuromusculares/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Alcohol Deshidrogenasa , Animales , Apoptosis/genética , Atrofia , Autofagia/genética , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Glutatión Reductasa/genética , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Factor 2 Relacionado con NF-E2/metabolismo , Óxido Nítrico/metabolismo , Oxidación-Reducción , Regeneración/genética , Tirosina/metabolismo
14.
Int J Cell Biol ; 2014: 428764, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24627685

RESUMEN

Protein S-nitrosation is deemed as a prototype of posttranslational modifications governing cell signaling. It takes place on specific cysteine residues that covalently incorporate a nitric oxide (NO) moiety to form S-nitrosothiol derivatives and depends on the ratio between NO produced by NO synthases and nitrosothiol removal catalyzed by denitrosating enzymes. A large number of cysteine-containing proteins are found to undergo S-nitrosation and, among them, the enzymes catalyzing ubiquitination, mainly the class of ubiquitin E3 ligases and the 20S component of the proteasome, have been reported to be redox modulated in their activity. In this review we will outline the processes regulating S-nitrosation and try to debate whether and how it affects protein ubiquitination and degradation via the proteasome. In particular, since muscle and neuronal health largely depends on the balance between protein synthesis and breakdown, here we will discuss the impact of S-nitrosation in the efficiency of protein quality control system, providing lines of evidence and speculating about its involvement in the onset and maintenance of neuromuscular dysfunctions.

15.
Int J Cell Biol ; 2012: 361872, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22927857

RESUMEN

S-nitrosylation is a posttranslational modification of cysteine residues that has been frequently indicated as potential molecular mechanism governing cell response upon redox unbalance downstream of nitric oxide (over)production. In the last years, increased levels of S-nitrosothiols (SNOs) have been tightly associated with the onset of nitroxidative stress-based pathologies (e.g., cancer and neurodegeneration), conditions in which alterations of mitochondrial homeostasis and activation of cellular processes dependent on it have been reported as well. In this paper we aim at summarizing the current knowledge of mitochondria-related proteins undergoing S-nitrosylation and how this redox modification might impact on mitochondrial functions, whose impairment has been correlated to tumorigenesis and neuronal cell death. In particular, emphasis will be given to the possible, but still neglected implication of denitrosylation reactions in the modulation of mitochondrial SNOs and how they can affect mitochondrion-related cellular process, such as oxidative phosphorylation, mitochondrial dynamics, and mitophagy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...