Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Res Sq ; 2023 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-37502961

RESUMEN

The uptake of Ca2+ into and extrusion of calcium from the mitochondrial matrix, regulated by the mitochondrial Ca2+ uniporter (MCU), is a fundamental biological process that has crucial impacts on cellular metabolism, signaling, growth and survival. Herein, we report that the embryonic lethality of Mcu-deficient mice is fully rescued by orally supplementing ferroptosis inhibitor lipophilic antioxidant vitamin E and ubiquinol. Mechanistically, we found MCU promotes acetyl-CoA-mediated GPX4 acetylation at K90 residue, and K90R mutation impaired the GPX4 enzymatic activity, a step that is crucial for ferroptosis. Structural analysis supports the possibility that GPX4 K90R mutation alters the conformational state of the molecule, resulting in disruption of a salt bridge formation with D23, which was confirmed by mutagenesis studies. Finally, we report that deletion of MCU in cancer cells caused a marked reduction in tumor growth in multiple cancer models. In summary, our study provides a first direct link between mitochondrial calcium level and sustained GPX4 enzymatic activity to regulate ferroptosis, which consequently protects cancer cells from ferroptosis.

2.
Biochem Soc Trans ; 50(3): 1205-1213, 2022 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-35758268

RESUMEN

GPX4 is a selenocysteine-containing protein that plays an essential role in repairing peroxidised phospholipids. Its role in organismal homeostasis has been known for decades, and it has been reported to play a pivotal role in cell survival and mammalian embryonic development. In recent years, GPX4 has been associated with a cell death modality dubbed ferroptosis. The framing of this molecular pathway of cell death was essential for understanding the conditions that determine GPX4 dependency and ultimately to the process of lipid peroxidation. Since its discovery, ferroptosis has been gaining momentum as a promising target for yet-incurable diseases, including cancer and neurodegeneration. Given the current interest, in the present review, we provide newcomers in the field with an overview of the biology of GPX4 and cover some of its most recent discoveries.


Asunto(s)
Ferroptosis , Animales , Muerte Celular , Glutatión Peroxidasa/metabolismo , Peroxidación de Lípido/fisiología , Mamíferos/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa
3.
Nat Commun ; 13(1): 1046, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35210435

RESUMEN

The most common cause of death in the intensive care unit (ICU) is the development of multiorgan dysfunction syndrome (MODS). Besides life-supporting treatments, no cure exists, and its mechanisms are still poorly understood. Catalytic iron is associated with ICU mortality and is known to cause free radical-mediated cellular toxicity. It is thought to induce excessive lipid peroxidation, the main characteristic of an iron-dependent type of cell death conceptualized as ferroptosis. Here we show that the severity of multiorgan dysfunction and the probability of death are indeed associated with plasma catalytic iron and lipid peroxidation. Transgenic approaches underscore the role of ferroptosis in iron-induced multiorgan dysfunction. Blocking lipid peroxidation with our highly soluble ferrostatin-analogue protects mice from injury and death in experimental non-septic multiorgan dysfunction, but not in sepsis-induced multiorgan dysfunction. The limitations of the experimental mice models to mimic the complexity of clinical MODS warrant further preclinical testing. In conclusion, our data suggest ferroptosis targeting as possible treatment option for a stratifiable subset of MODS patients.


Asunto(s)
Ferroptosis , Animales , Muerte Celular , Humanos , Hierro/metabolismo , Peroxidación de Lípido , Ratones , Insuficiencia Multiorgánica/prevención & control
4.
Redox Biol ; 48: 102188, 2021 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-34794077

RESUMEN

Selenoproteins are a small family of proteins containing the trace element selenium in form of the rare amino acid selenocysteine (Sec), which is decoded by the UGA codon. In humans, a number of pathogenic variants in genes encoding distinct selenoproteins or selenoprotein biosynthesis factors have been identified. Pathogenic variants in selenocysteine synthase (SEPSECS), which catalyzes the last step in Sec-tRNA[Ser]Sec biosynthesis, were reported in children suffering from progressive cerebello-cerebral atrophy. To understand the pathomechanism associated with SEPSECS deficiency, we generated a novel mouse model recapitulating the respective human pathogenic p.Y334C variant in the murine Sepsecs gene (SepsecsY334C). Unlike in patients, pups homozygous for the p.Y334C variant died perinatally with signs of cardio-respiratory failure. Perinatal death is reminiscent of the Sedaghatian spondylometaphyseal dysplasia disorder in humans, which is caused by pathogenic variants in the gene encoding the selenoprotein and key ferroptosis regulator glutathione peroxidase 4 (GPX4). Protein expression levels of distinct selenoproteins in SepsecsY334C/Y334C mice were found to be generally reduced in brain and isolated cortical neurons, while transcriptomics analysis uncovered an upregulation of NRF2-regulated genes. Crossbreeding of SepsecsY334C/Y334C mice with mice harboring a targeted mutation of the catalytically active Sec to Cys in GPX4 rescued perinatal death of SepsecsY334C/Y334C mice, showing that the cardio-respiratory defects of SepsecsY334C/Y334C mice were caused by the lack of GPX4. Like in SepsecsY334C/Y334C mice, selenoprotein expression levels remained low and NRF2-regulated genes remained highly expressed in these compound mutant mice, indicating that selenium-independent GPX4, along with a sustained antioxidant response are sufficient to compensate for dysfunctional Sec-tRNA[Ser]Sec biosynthesis. Our findings imply that children with pathogenic variants in SEPSECS or GPX4 may even benefit from treatments that incompletely compensate for impaired GPX4 activity.

5.
Nat Commun ; 12(1): 4402, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34285231

RESUMEN

Acute kidney injury (AKI) is morphologically characterized by a synchronized plasma membrane rupture of cells in a specific section of a nephron, referred to as acute tubular necrosis (ATN). Whereas the involvement of necroptosis is well characterized, genetic evidence supporting the contribution of ferroptosis is lacking. Here, we demonstrate that the loss of ferroptosis suppressor protein 1 (Fsp1) or the targeted manipulation of the active center of the selenoprotein glutathione peroxidase 4 (Gpx4cys/-) sensitize kidneys to tubular ferroptosis, resulting in a unique morphological pattern of tubular necrosis. Given the unmet medical need to clinically inhibit AKI, we generated a combined small molecule inhibitor (Nec-1f) that simultaneously targets receptor interacting protein kinase 1 (RIPK1) and ferroptosis in cell lines, in freshly isolated primary kidney tubules and in mouse models of cardiac transplantation and of AKI and improved survival in models of ischemia-reperfusion injury. Based on genetic and pharmacological evidence, we conclude that GPX4 dysfunction hypersensitizes mice to ATN during AKI. Additionally, we introduce Nec-1f, a solid inhibitor of RIPK1 and weak inhibitor of ferroptosis.


Asunto(s)
Lesión Renal Aguda/patología , Ferroptosis/fisiología , Túbulos Renales/patología , Daño por Reperfusión/patología , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/etiología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Cisplatino/administración & dosificación , Cisplatino/toxicidad , Modelos Animales de Enfermedad , Células Epiteliales , Femenino , Ferroptosis/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células HT29 , Trasplante de Corazón/efectos adversos , Humanos , Imidazoles/química , Imidazoles/farmacología , Imidazoles/uso terapéutico , Indoles/química , Indoles/farmacología , Indoles/uso terapéutico , Masculino , Ratones , Ratones Transgénicos , Microsomas Hepáticos , Proteínas Mitocondriales/metabolismo , Células 3T3 NIH , Necrosis/tratamiento farmacológico , Necrosis/etiología , Necrosis/patología , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/antagonistas & inhibidores , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Cultivo Primario de Células , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/etiología
6.
Nature ; 575(7784): 693-698, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31634899

RESUMEN

Ferroptosis is an iron-dependent form of necrotic cell death marked by oxidative damage to phospholipids1,2. To date, ferroptosis has been thought to be controlled only by the phospholipid hydroperoxide-reducing enzyme glutathione peroxidase 4 (GPX4)3,4 and radical-trapping antioxidants5,6. However, elucidation of the factors that underlie the sensitivity of a given cell type to ferroptosis7 is crucial to understand the pathophysiological role of ferroptosis and how it may be exploited for the treatment of cancer. Although metabolic constraints8 and phospholipid composition9,10 contribute to ferroptosis sensitivity, no cell-autonomous mechanisms have been identified that account for the resistance of cells to ferroptosis. Here we used an expression cloning approach to identify genes in human cancer cells that are able to complement the loss of GPX4. We found that the flavoprotein apoptosis-inducing factor mitochondria-associated 2 (AIFM2) is a previously unrecognized anti-ferroptotic gene. AIFM2, which we renamed ferroptosis suppressor protein 1 (FSP1) and which was initially described as a pro-apoptotic gene11, confers protection against ferroptosis elicited by GPX4 deletion. We further demonstrate that the suppression of ferroptosis by FSP1 is mediated by ubiquinone (also known as coenzyme Q10, CoQ10): the reduced form, ubiquinol, traps lipid peroxyl radicals that mediate lipid peroxidation, whereas FSP1 catalyses the regeneration of CoQ10 using NAD(P)H. Pharmacological targeting of FSP1 strongly synergizes with GPX4 inhibitors to trigger ferroptosis in a number of cancer entities. In conclusion, the FSP1-CoQ10-NAD(P)H pathway exists as a stand-alone parallel system, which co-operates with GPX4 and glutathione to suppress phospholipid peroxidation and ferroptosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Ferroptosis/genética , Glutatión/metabolismo , Proteínas Mitocondriales/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Peroxidación de Lípido/genética , Ratones , Proteínas Mitocondriales/genética , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo
7.
J Clin Invest ; 128(8): 3341-3355, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29939160

RESUMEN

High-risk neuroblastoma is a devastating malignancy with very limited therapeutic options. Here, we identify withaferin A (WA) as a natural ferroptosis-inducing agent in neuroblastoma, which acts through a novel double-edged mechanism. WA dose-dependently either activates the nuclear factor-like 2 pathway through targeting of Kelch-like ECH-associated protein 1 (noncanonical ferroptosis induction) or inactivates glutathione peroxidase 4 (canonical ferroptosis induction). Noncanonical ferroptosis induction is characterized by an increase in intracellular labile Fe(II) upon excessive activation of heme oxygenase-1, which is sufficient to induce ferroptosis. This double-edged mechanism might explain the superior efficacy of WA as compared with etoposide or cisplatin in killing a heterogeneous panel of high-risk neuroblastoma cells, and in suppressing the growth and relapse rate of neuroblastoma xenografts. Nano-targeting of WA allows systemic application and suppressed tumor growth due to an enhanced accumulation at the tumor site. Collectively, our data propose a novel therapeutic strategy to efficiently kill cancer cells by ferroptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Neuroblastoma/tratamiento farmacológico , Witanólidos/farmacología , Animales , Línea Celular Tumoral , Embrión de Pollo , Hemo-Oxigenasa 1/metabolismo , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Ratones , Proteínas de Neoplasias/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Oncotarget ; 9(32): 22241-22242, 2018 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-29854274
9.
Cell ; 172(3): 409-422.e21, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29290465

RESUMEN

Selenoproteins are rare proteins among all kingdoms of life containing the 21st amino acid, selenocysteine. Selenocysteine resembles cysteine, differing only by the substitution of selenium for sulfur. Yet the actual advantage of selenolate- versus thiolate-based catalysis has remained enigmatic, as most of the known selenoproteins also exist as cysteine-containing homologs. Here, we demonstrate that selenolate-based catalysis of the essential mammalian selenoprotein GPX4 is unexpectedly dispensable for normal embryogenesis. Yet the survival of a specific type of interneurons emerges to exclusively depend on selenocysteine-containing GPX4, thereby preventing fatal epileptic seizures. Mechanistically, selenocysteine utilization by GPX4 confers exquisite resistance to irreversible overoxidation as cells expressing a cysteine variant are highly sensitive toward peroxide-induced ferroptosis. Remarkably, concomitant deletion of all selenoproteins in Gpx4cys/cys cells revealed that selenoproteins are dispensable for cell viability provided partial GPX4 activity is retained. Conclusively, 200 years after its discovery, a specific and indispensable role for selenium is provided.


Asunto(s)
Apoptosis , Glutatión Peroxidasa/metabolismo , Convulsiones/metabolismo , Selenio/metabolismo , Animales , Supervivencia Celular , Células Cultivadas , Femenino , Glutatión Peroxidasa/genética , Células HEK293 , Humanos , Peróxido de Hidrógeno/toxicidad , Interneuronas/metabolismo , Peroxidación de Lípido , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Convulsiones/etiología
10.
Nat Chem Biol ; 13(1): 91-98, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27842070

RESUMEN

Ferroptosis is a form of regulated necrotic cell death controlled by glutathione peroxidase 4 (GPX4). At present, mechanisms that could predict sensitivity and/or resistance and that may be exploited to modulate ferroptosis are needed. We applied two independent approaches-a genome-wide CRISPR-based genetic screen and microarray analysis of ferroptosis-resistant cell lines-to uncover acyl-CoA synthetase long-chain family member 4 (ACSL4) as an essential component for ferroptosis execution. Specifically, Gpx4-Acsl4 double-knockout cells showed marked resistance to ferroptosis. Mechanistically, ACSL4 enriched cellular membranes with long polyunsaturated ω6 fatty acids. Moreover, ACSL4 was preferentially expressed in a panel of basal-like breast cancer cell lines and predicted their sensitivity to ferroptosis. Pharmacological targeting of ACSL4 with thiazolidinediones, a class of antidiabetic compound, ameliorated tissue demise in a mouse model of ferroptosis, suggesting that ACSL4 inhibition is a viable therapeutic approach to preventing ferroptosis-related diseases.


Asunto(s)
Apoptosis , Coenzima A Ligasas/metabolismo , Glutatión Peroxidasa/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Coenzima A Ligasas/antagonistas & inhibidores , Coenzima A Ligasas/deficiencia , Femenino , Glutatión Peroxidasa/deficiencia , Humanos , Hipoglucemiantes/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Noqueados , Necrosis , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Tiazolidinedionas/farmacología
11.
Nat Nanotechnol ; 11(11): 977-985, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27668796

RESUMEN

The design of cancer-targeting particles with precisely tuned physicochemical properties may enhance the delivery of therapeutics and access to pharmacological targets. However, a molecular-level understanding of the interactions driving the fate of nanomedicine in biological systems remains elusive. Here, we show that ultrasmall (<10 nm in diameter) poly(ethylene glycol)-coated silica nanoparticles, functionalized with melanoma-targeting peptides, can induce a form of programmed cell death known as ferroptosis in starved cancer cells and cancer-bearing mice. Tumour xenografts in mice intravenously injected with nanoparticles using a high-dose multiple injection scheme exhibit reduced growth or regression, in a manner that is reversed by the pharmacological inhibitor of ferroptosis, liproxstatin-1. These data demonstrate that ferroptosis can be targeted by ultrasmall silica nanoparticles and may have therapeutic potential.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Hierro/metabolismo , Nanopartículas/química , Aminoácidos/deficiencia , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Lisosomas/efectos de los fármacos , Melanoma , Ratones , Ratones SCID , Nanopartículas/uso terapéutico , Tamaño de la Partícula , Polietilenglicoles/química , Quinoxalinas/farmacología , Dióxido de Silicio/química , Compuestos de Espiro/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , alfa-MSH/química
12.
PLoS Genet ; 12(1): e1005800, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26808544

RESUMEN

The tripeptide glutathione is the most abundant cellular antioxidant with high medical relevance, and it is also required as a co-factor for various enzymes involved in the detoxification of reactive oxygen species and toxic compounds. However, its cell-type specific functions and its interaction with other cytoprotective molecules are largely unknown. Using a combination of mouse genetics, functional cell biology and pharmacology, we unraveled the function of glutathione in keratinocytes and its cross-talk with other antioxidant defense systems. Mice with keratinocyte-specific deficiency in glutamate cysteine ligase, which catalyzes the rate-limiting step in glutathione biosynthesis, showed a strong reduction in keratinocyte viability in vitro and in the skin in vivo. The cells died predominantly by apoptosis, but also showed features of ferroptosis and necroptosis. The increased cell death was associated with increased levels of reactive oxygen and nitrogen species, which caused DNA and mitochondrial damage. However, epidermal architecture, and even healing of excisional skin wounds were only mildly affected in the mutant mice. The cytoprotective transcription factor Nrf2 was strongly activated in glutathione-deficient keratinocytes, but additional loss of Nrf2 did not aggravate the phenotype, demonstrating that the cytoprotective effect of Nrf2 is glutathione dependent. However, we show that deficiency in glutathione biosynthesis is efficiently compensated in keratinocytes by the cysteine/cystine and thioredoxin systems. Therefore, our study highlights a remarkable antioxidant capacity of the epidermis that ensures skin integrity and efficient wound healing.


Asunto(s)
Glutatión/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Piel/metabolismo , Tiorredoxinas/genética , Animales , Antioxidantes/metabolismo , Apoptosis/genética , Supervivencia Celular , Daño del ADN/genética , Glutamato-Cisteína Ligasa/deficiencia , Glutamato-Cisteína Ligasa/genética , Glutamato-Cisteína Ligasa/metabolismo , Glutatión/biosíntesis , Queratinocitos/metabolismo , Queratinocitos/patología , Ratones , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Piel/patología , Tiorredoxinas/metabolismo , Cicatrización de Heridas/genética
13.
Brain ; 139(Pt 2): 346-54, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26626369

RESUMEN

Thioredoxin 2 (TXN2; also known as Trx2) is a small mitochondrial redox protein essential for the control of mitochondrial reactive oxygen species homeostasis, apoptosis regulation and cell viability. Exome sequencing in a 16-year-old adolescent suffering from an infantile-onset neurodegenerative disorder with severe cerebellar atrophy, epilepsy, dystonia, optic atrophy, and peripheral neuropathy, uncovered a homozygous stop mutation in TXN2. Analysis of patient-derived fibroblasts demonstrated absence of TXN2 protein, increased reactive oxygen species levels, impaired oxidative stress defence and oxidative phosphorylation dysfunction. Reconstitution of TXN2 expression restored all these parameters, indicating the causal role of TXN2 mutation in disease development. Supplementation with antioxidants effectively suppressed cellular reactive oxygen species production, improved cell viability and mitigated clinical symptoms during short-term follow-up. In conclusion, our report on a patient with TXN2 deficiency suggests an important role of reactive oxygen species homeostasis for human neuronal maintenance and energy metabolism.


Asunto(s)
Homeostasis/fisiología , Mitocondrias/metabolismo , Proteínas Mitocondriales/deficiencia , Enfermedades Neurodegenerativas/diagnóstico , Enfermedades Neurodegenerativas/metabolismo , Niño , Humanos , Masculino , Mitocondrias/genética , Proteínas Mitocondriales/genética , Enfermedades Neurodegenerativas/genética , Oxidación-Reducción , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Tiorredoxinas/genética
14.
J Biol Chem ; 290(23): 14668-78, 2015 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-25922076

RESUMEN

The selenoenzyme Gpx4 is essential for early embryogenesis and cell viability for its unique function to prevent phospholipid oxidation. Recently, the cytosolic form of Gpx4 was identified as an upstream regulator of a novel form of non-apoptotic cell death, called ferroptosis, whereas the mitochondrial isoform of Gpx4 was previously shown to be crucial for male fertility. Here, we generated and analyzed mice with a targeted mutation of the active site selenocysteine of Gpx4 (Gpx4_U46S). Mice homozygous for Gpx4_U46S died at the same embryonic stage (E7.5) as Gpx4(-/-) embryos as expected. Surprisingly, male mice heterozygous for Gpx4_U46S presented subfertility. Subfertility was manifested in a reduced number of litters from heterozygous breeding and an impairment of spermatozoa to fertilize oocytes in vitro. Morphologically, sperm isolated from heterozygous Gpx4_U46S mice revealed many structural abnormalities particularly in the spermatozoa midpiece due to improper oxidation and polymerization of sperm capsular proteins and malformation of the mitochondrial capsule surrounding and stabilizing sperm mitochondria. These findings are reminiscent of sperm isolated from selenium-deprived rodents or from mice specifically lacking mitochondrial Gpx4. Due to a strongly facilitated incorporation of Ser in the polypeptide chain as compared with selenocysteine at the UGA codon, expression of the catalytically inactive Gpx4_U46S was found to be strongly increased. Because the stability of the mitochondrial capsule of mature spermatozoa depends on the moonlighting function of Gpx4 both as an enzyme oxidizing capsular protein thiols and as a structural protein, tightly controlled expression of functional Gpx4 emerges as a key for full male fertility.


Asunto(s)
Sustitución de Aminoácidos , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Infertilidad Masculina/genética , Espermatogénesis , Animales , Dominio Catalítico , Células Cultivadas , Pérdida del Embrión/genética , Pérdida del Embrión/metabolismo , Pérdida del Embrión/patología , Femenino , Heterocigoto , Homocigoto , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Masculino , Ratones , Ratones Transgénicos , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Selenocisteína/genética , Serina/genética , Espermatozoides/metabolismo , Espermatozoides/patología , Espermatozoides/ultraestructura
15.
Antioxid Redox Signal ; 22(11): 938-50, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25647640

RESUMEN

AIMS: Mitochondrial thioredoxin reductase (Txnrd2) is a central player in the control of mitochondrial hydrogen peroxide (H2O2) abundance by serving as a direct electron donor to the thioredoxin-peroxiredoxin axis. In this study, we investigated the impact of targeted disruption of Txnrd2 on tumor growth. RESULTS: Tumor cells with a Txnrd2 deficiency failed to activate hypoxia-inducible factor-1α (Hif-1α) signaling; it rather caused PHD2 accumulation, Hif-1α degradation and decreased vascular endothelial growth factor (VEGF) levels, ultimately leading to reduced tumor growth and tumor vascularization. Increased c-Jun NH2-terminal Kinase (JNK) activation proved to be the molecular link between the loss of Txnrd2, an altered mitochondrial redox balance with compensatory upregulation of glutaredoxin-2, and elevated PHD2 expression. INNOVATION: Our data provide compelling evidence for a yet-unrecognized mitochondrial Txnrd-driven, regulatory mechanism that ultimately prevents cellular Hif-1α accumulation. In addition, simultaneous targeting of both the mitochondrial thioredoxin and glutathione systems was used as an efficient therapeutic approach in hindering tumor growth. CONCLUSION: This work demonstrates an unexpected regulatory link between mitochondrial Txnrd and the JNK-PHD2-Hif-1α axis, which highlights how the loss of Txnrd2 and the resulting altered mitochondrial redox balance impairs tumor growth as well as tumor-related angiogenesis. Furthermore, it opens a new avenue for a therapeutic approach to hinder tumor growth by the simultaneous targeting of both the mitochondrial thioredoxin and glutathione systems.


Asunto(s)
Proliferación Celular , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Mitocondrias/metabolismo , Neovascularización Patológica/metabolismo , Tiorredoxina Reductasa 2/genética , Animales , Células Cultivadas , Técnicas de Silenciamiento del Gen , Xenoinjertos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Especies Reactivas de Oxígeno/metabolismo
16.
Biochim Biophys Acta ; 1850(8): 1566-74, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25450170

RESUMEN

BACKGROUND: During maturation and storage, spermatozoa generate substantial amounts of reactive oxygen species (ROS) and are thus forced to cope with an increasingly oxidative environment that is both needed and detrimental to their biology. Such a janus-faceted intermediate needs to be tightly controlled and this is done by a wide array of redox enzymes. These enzymes not only have to prevent unspecific modifications of essential cellular biomolecules by quenching undesired ROS, but they are also required and often directly involved in critical protein modifications. SCOPE OF REVIEW: The present review is conceived to present an update on what is known about critical roles of redox enzymes, whereby special emphasis is put on the family of glutathione peroxidases, which for the time being presents the best characterized tasks during gametogenesis. MAJOR CONCLUSIONS: We therefore demonstrate that understanding the function of (seleno)thiol-based oxidases/reductases is not a trivial task and relevant knowledge will be mainly gained by using robust systems, as exemplified by several (conditional) knockout studies. We thus stress the importance of using such models for providing unequivocal evidence in the molecular understanding of redox regulatory mechanisms in sperm maturation. GENERAL SIGNIFICANCE: ROS are not merely detrimental by-products of metabolism and their proper generation and usage by specific enzymes is essential for vital functions as beautifully exemplified during male gametogenesis. As such, lessons learnt from thiol-based oxidases/reductases in male gametogenesis could be used as a general principle for other organs as it is most likely not only restricted to this developmental phase. This article is part of a Special Issue entitled Redox regulation of differentiation and de-differentiation.


Asunto(s)
Especies Reactivas de Oxígeno/metabolismo , Espermatogénesis , Espermatozoides/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Animales , Glutatión Peroxidasa/metabolismo , Humanos , Masculino , Modelos Biológicos , Oxidación-Reducción , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Espermatozoides/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...