Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 10(38): eadj4122, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39303039

RESUMEN

Sarcopenia is characterized by accelerated muscle mass and function loss, which burdens and challenges public health worldwide. Several studies indicated that selenium deficiency is associated with sarcopenia; however, the specific mechanism remains unclear. Here, we demonstrated that selenoprotein W (SELENOW) containing selenium in the form of selenocysteine functioned in sarcopenia. SELENOW expression is up-regulated in dexamethasone (DEX)-induced muscle atrophy and age-related sarcopenia mouse models. Knockout (KO) of SELENOW profoundly aggravated the process of muscle mass loss in the two mouse models. Mechanistically, SELENOW KO suppressed the RAC1-mTOR cascade by the interaction between SELENOW and RAC1 and induced the imbalance of protein synthesis and degradation. Consistently, overexpression of SELENOW in vivo and in vitro alleviated the muscle and myotube atrophy induced by DEX. SELENOW played a role in age-related sarcopenia and regulated the genes associated with aging. Together, our study uncovered the function of SELENOW in age-related sarcopenia and provides promising evidence for the prevention and treatment of sarcopenia.


Asunto(s)
Ratones Noqueados , Complejo de la Endopetidasa Proteasomal , Biosíntesis de Proteínas , Sarcopenia , Selenoproteína W , Ubiquitina , Animales , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratones , Sarcopenia/metabolismo , Sarcopenia/genética , Sarcopenia/patología , Ubiquitina/metabolismo , Selenoproteína W/genética , Selenoproteína W/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/efectos de los fármacos , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/genética , Dexametasona/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Modelos Animales de Enfermedad , Atrofia Muscular/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/patología , Atrofia Muscular/inducido químicamente , Envejecimiento/metabolismo , Masculino , Transducción de Señal , Neuropéptidos
2.
Commun Biol ; 7(1): 872, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39020075

RESUMEN

Lower selenium levels are observed in Alzheimer's disease (AD) brains, while supplementation shows multiple benefits. Selenoprotein W (SELENOW) is sensitive to selenium changes and binds to tau, reducing tau accumulation. However, whether restoration of SELENOW has any protective effect in AD models and its underlying mechanism remain unknown. Here, we confirm the association between SELENOW downregulation and tau pathology, revealing SELENOW's role in promoting tau degradation through the ubiquitin‒proteasome system. SELENOW competes with Hsp70 to interact with tau, promoting its ubiquitination and inhibiting tau acetylation at K281. SELENOW deficiency leads to synaptic defects, tau dysregulation and impaired long-term potentiation, resulting in memory deficits in mice. Conversely, SELENOW overexpression in the triple transgenic AD mice ameliorates memory impairment and tau-related pathologies, featuring decreased 4-repeat tau isoform, phosphorylation at Ser396 and Ser404, neurofibrillary tangles and neuroinflammation. Thus, SELENOW contributes to the regulation of tau homeostasis and synaptic maintenance, implicating its potential role in AD.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Homeostasis , Ratones Transgénicos , Selenoproteína W , Proteínas tau , Animales , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Proteínas tau/metabolismo , Proteínas tau/genética , Ratones , Selenoproteína W/metabolismo , Selenoproteína W/genética , Masculino , Fosforilación , Humanos , Ratones Endogámicos C57BL
3.
Mol Biol Rep ; 51(1): 587, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38683225

RESUMEN

BACKGROUND: Patients with multiple myeloma exhibit malignant osteolytic bone disease due to excessive osteoclast formation and function. We recently identified that osteoclastogenic stimulator selenoprotein W (SELENOW) is upregulated via ERK signaling and downregulated via p38 signaling during receptor activator of nuclear factor (NF)-κΒ ligand (RANKL)-induced osteoclast differentiation. In the intrinsic physiological process, RANKL-induced downregulation of SELENOW maintains proper osteoclast differentiation; in contrast, forced overexpression of SELENOW leads to overactive osteoclast formation and function. METHODS AND RESULTS: We observed that SELENOW is highly expressed in multiple myeloma-derived peripheral blood mononuclear cells (PBMCs) and mature osteoclasts when compared to healthy controls. Also, the level of tumor necrosis factor alpha (TNFα), a pathological osteoclastogenic factor, is increased in the PBMCs and serum of patients with multiple myeloma. ERK activation by TNFα was more marked and sustained than that by RANKL, allowing SELENOW upregulation. Excessive expression of SELENOW in osteoclast progenitors and mature osteoclasts derived from multiple myeloma facilitated efficient nuclear translocation of osteoclastogenic transcription factors NF-κB and NFATc1, which are favorable for osteoclast formation. CONCLUSION: Our findings suggest a possibility that feedforward signaling of osteoclastogenic SELENOW by TNFα derived from multiple myeloma induces overactive osteoclast differentiation, leading to bone loss during multiple myeloma.


Asunto(s)
Diferenciación Celular , Mieloma Múltiple , Osteoclastos , Selenoproteína W , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Diferenciación Celular/genética , Leucocitos Mononucleares/metabolismo , Sistema de Señalización de MAP Quinasas , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Mieloma Múltiple/genética , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Factores de Transcripción NFATC/genética , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Selenoproteína W/metabolismo , Selenoproteína W/genética , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
4.
Redox Biol ; 71: 103114, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38460355

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease worldwide. Numerous evidence has demonstrated that metabolic reprogramming serves as a hallmark associated with an elevated risk of NAFLD progression. Selenoprotein W (SelW) is an extensively expressed hepatic selenoprotein that plays a crucial role in antioxidant function. Here, we first demonstrated that SelW is a significantly distinct factor in the liver tissue of NAFLD patients through the Gene Expression Omnibus (GEO) database. Additionally, loss of SelW alleviated hepatic steatosis induced by a high-fat diet (HFD), and was accompanied by the regulation of metabolic and inflammatory pathways as verified by transcriptomic analysis. Moreover, co-immunoprecipitation (CO-IP), liquid chromatography-tandem mass spectrometry (LC-MS), laser scanning confocal microscopy (LSCM) and molecular docking analysis were subsequently implemented to identify Pyruvate Kinase M2 (PKM2) as a potential interacting protein of SelW. Meanwhile, SelW modulated PKM2 translocation into the nucleus to trigger transactivation of the HIF-1α, in further mediating mitochondrial apoptosis, eventually resulting in mitochondrial damage, ROS excessive production and mtDNA leakage. Additionally, mito-ROS accumulation induced the activation of the NLRP3 inflammasome-mediated pyroptosis, thereby facilitating extracellular leakage of mtDNA. The escaped mtDNA then evokes the cGAS-STING signaling pathway in macrophage, thus inducing a shift in macrophage phenotype. Together, our results suggest SelW promotes hepatocyte apoptosis and pyroptosis by regulating metabolic reprogramming to activate cGAS/STING signaling of macrophages, thereby exacerbating the progression of NAFLD.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Animales , Humanos , Ratones , Dieta Alta en Grasa , ADN Mitocondrial/metabolismo , Hígado/metabolismo , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Nucleotidiltransferasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Selenoproteína W/metabolismo
5.
Zhonghua Liu Xing Bing Xue Za Zhi ; 44(4): 629-635, 2023 Apr 10.
Artículo en Chino | MEDLINE | ID: mdl-37147837

RESUMEN

Objective: The docking and superantigen activity sites of staphylococcal enterotoxin-like W (SElW) and T cell receptor (TCR) were predicted, and its SElW was cloned, expressed and purified. Methods: AlphaFold was used to predict the 3D structure of SElW protein monomers, and the protein models were evaluated with the help of the SAVES online server from ERRAT, Ramachandran plot, and Verify_3D. The ZDOCK server simulates the docking conformation of SElW and TCR, and the amino acid sequences of SElW and other serotype enterotoxins were aligned. The primers were designed to amplify selw, and the fragment was recombined into the pMD18-T vector and sequenced. Then recombinant plasmid pMD18-T was digested with BamHⅠand Hind Ⅲ. The target fragment was recombined into the expression plasmid pET-28a(+). After identification of the recombinant plasmid, the protein expression was induced by isopropyl-beta-D- thiogalactopyranoside. The SElW expressed in the supernatant was purified by affinity chromatography and quantified by the BCA method. Results: The predicted three-dimensional structure showed that the SElW protein was composed of two domains, the amino-terminal and the carboxy-terminal. The amino-terminal domain was composed of 3 α-helices and 6 ß-sheets, and the carboxy-terminal domain included 2 α-helices and 7 antiparallel ß-sheets composition. The overall quality factor score of the SElW protein model was 98.08, with 93.24% of the amino acids having a Verify_3D score ≥0.2 and no amino acids located in disallowed regions. The docking conformation with the highest score (1 521.328) was selected as the analysis object, and the 19 hydrogen bonds between the corresponding amino acid residues of SElW and TCR were analyzed by PyMOL. Combined with sequence alignment and the published data, this study predicted and found five important superantigen active sites, namely Y18, N19, W55, C88, and C98. The highly purified soluble recombinant protein SElW was obtained with cloning, expression, and protein purification. Conclusions: The study found five superantigen active sites in SElW protein that need special attention and successfully constructed and expressed the SElW protein, which laid the foundation for further exploration of the immune recognition mechanism of SElW.


Asunto(s)
Enterotoxinas , Superantígenos , Humanos , Enterotoxinas/genética , Superantígenos/genética , Dominio Catalítico , Selenoproteína W/metabolismo , Receptores de Antígenos de Linfocitos T
6.
Redox Biol ; 59: 102571, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36516721

RESUMEN

Macrophages play a pivotal role in mediating inflammation and subsequent resolution of inflammation. The availability of selenium as a micronutrient and the subsequent biosynthesis of selenoproteins, containing the 21st amino acid selenocysteine (Sec), are important for the physiological functions of macrophages. Selenoproteins regulate the redox tone in macrophages during inflammation, the early onset of which involves oxidative burst of reactive oxygen and nitrogen species. SELENOW is a highly expressed selenoprotein in bone marrow-derived macrophages (BMDMs). Beyond its described general role as a thiol and peroxide reductase and as an interacting partner for 14-3-3 proteins, its cellular functions, particularly in macrophages, remain largely unknown. In this study, we utilized Selenow knock-out (KO) murine bone marrow-derived macrophages (BMDMs) to address the role of SELENOW in inflammation following stimulation with bacterial endotoxin lipopolysaccharide (LPS). RNAseq-based temporal analyses of expression of selenoproteins and the Sec incorporation machinery genes suggested no major differences in the selenium utilization pathway in the Selenow KO BMDMs compared to their wild-type counterparts. However, selective enrichment of oxidative stress-related selenoproteins and increased ROS in Selenow-/- BMDMs indicated anomalies in redox homeostasis associated with hierarchical expression of selenoproteins. Selenow-/- BMDMs also exhibited reduced expression of arginase-1, a key enzyme associated with anti-inflammatory (M2) phenotype necessary to resolve inflammation, along with a significant decrease in efferocytosis of neutrophils that triggers pathways of resolution. Parallel targeted metabolomics analysis also confirmed an impairment in arginine metabolism in Selenow-/- BMDMs. Furthermore, Selenow-/- BMDMs lacked the ability to enhance characteristic glycolytic metabolism during inflammation. Instead, these macrophages atypically relied on oxidative phosphorylation for energy production when glucose was used as an energy source. These findings suggest that SELENOW expression in macrophages may have important implications on cellular redox processes and bioenergetics during inflammation and its resolution.


Asunto(s)
Selenio , Selenoproteína W , Ratones , Animales , Selenoproteína W/genética , Selenoproteína W/metabolismo , Selenio/metabolismo , Selenoproteínas/genética , Selenoproteínas/metabolismo , Macrófagos/metabolismo , Oxidación-Reducción , Inflamación/genética
7.
Biometals ; 35(6): 1359-1370, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36261677

RESUMEN

Selenium (Se) plays an essential role in the growth of fish and performs its physiological functions mainly through incorporation into selenoproteins. Our previous studies suggested that the selenoprotein W gene (selenow) is sensitive to changes in dietary Se in rainbow trout. However, the molecular characterization and tissue expression pattern of selenow are still unknown. Here, we revealed the molecular characterization, the tissue expression pattern of rainbow trout selenow and analyzed its response to dietary Se. The open reading frame (ORF) of the selenow gene was composed of 393 base pairs (bp) and encodes a 130-amino-acid protein. The 3' untranslated region (UTR) was 372 bp with a selenocysteine insertion sequence (SECIS) element. Remarkably, the rainbow trout selenow gene sequence was longer than those reported for mammals and most other fish. A ß1-α1-ß2-ß3-ß4-α2 pattern made up the secondary structure of SELENOW. Furthermore, multiple sequence alignment revealed that rainbow trout SELENOW showed a high level of identity with SELENOW from Salmo salar. In addition, the selenow gene was ubiquitously distributed in 13 tissues with various abundances and was predominantly expressed in muscle and brain. Interestingly, dietary Se significantly increased selenow mRNA expression in muscle. Our results highlight the vital role of selenow in rainbow trout muscle response to dietary Se levels and provide a theoretical basis for studies of selenow.


Asunto(s)
Oncorhynchus mykiss , Selenio , Animales , Oncorhynchus mykiss/genética , Oncorhynchus mykiss/metabolismo , Selenoproteína W/genética , Selenoproteína W/metabolismo , Selenio/metabolismo , Selenocisteína/genética , Selenocisteína/metabolismo , Selenoproteínas/genética , Selenoproteínas/metabolismo , Clonación Molecular , Mamíferos/genética
8.
Int J Mol Sci ; 22(19)2021 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-34638679

RESUMEN

Selenoprotein W (SELENOW) is a 9.6 kDa protein containing selenocysteine (Sec, U) in a conserved Cys-X-X-Sec (CXXU) motif. Previously, we reported that SELENOW regulates various cellular processes by interacting with 14-3-3ß at the U of the CXXU motif. Thioredoxin (Trx) is a small protein that plays a key role in the cellular redox regulatory system. The CXXC motif of Trx is critical for redox regulation. Recently, an interaction between Trx1 and 14-3-3 has been predicted. However, the binding mechanism and its biological effects remain unknown. In this study, we found that Trx1 interacted with 14-3-3ß at the Cys32 residue in the CXXC motif, and SELENOW and Trx1 were bound at Cys191 residue of 14-3-3ß. In vitro binding assays showed that SELENOW and Trx1 competed for interaction with 14-3-3ß. Compared to control cells, Trx1-deficient cells and SELENOW-deficient cells showed increased levels of both the subG1 population and poly (ADP-ribose) polymerase (PARP) cleavage by etoposide treatment. Moreover, Akt phosphorylation of Ser473 was reduced in Trx1-deficient cells and was recovered by overexpression of SELENOW. These results indicate that SELENOW can protect Trx1-deficient cells from etoposide-induced cell death through its interaction with 14-3-3ß.


Asunto(s)
Proteínas 14-3-3/metabolismo , Muerte Celular/efectos de los fármacos , Etopósido/farmacología , Selenoproteína W/farmacología , Tiorredoxinas/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Células HEK293 , Humanos , Células MCF-7 , Ratones , Oxidación-Reducción/efectos de los fármacos , Fosforilación/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
9.
Immunity ; 54(8): 1728-1744.e7, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34343498

RESUMEN

Inflammatory bowel disease (IBD) mainly includes Crohn's disease (CD) and ulcerative colitis (UC). Immune disorders play an essential role in the pathogenesis of these two IBDs, but the differences in the immune microenvironment of the colon and their underlying mechanisms remain poorly investigated. Here we examined the immunological features and metabolic microenvironment of untreated individuals with IBD by multiomics analyses. Modulation of CD-specific metabolites, particularly reduced selenium, can obviously shape type 1 T helper (Th1) cell differentiation, which is specifically enriched in CD. Selenium supplementation suppressed the symptoms and onset of CD and Th1 cell differentiation via selenoprotein W (SELW)-mediated cellular reactive oxygen species scavenging. SELW promoted purine salvage pathways and inhibited one-carbon metabolism by recruiting an E3 ubiquitin ligase, tripartite motif-containing protein 21, which controlled the stability of serine hydroxymethyltransferase 2. Our work highlights selenium as an essential regulator of T cell responses and potential therapeutic targets in CD.


Asunto(s)
Antioxidantes/farmacología , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/inmunología , Selenio/farmacología , Selenoproteína W/metabolismo , Células TH1/citología , Diferenciación Celular/inmunología , Polaridad Celular , Colon/inmunología , Colon/patología , Glicina Hidroximetiltransferasa/metabolismo , Humanos , Especies Reactivas de Oxígeno/metabolismo , Ribonucleoproteínas/metabolismo , Células TH1/inmunología , Ubiquitina-Proteína Ligasas/metabolismo
10.
Biochim Biophys Acta Proteins Proteom ; 1869(10): 140685, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34216797

RESUMEN

Selenoprotein W is widespread among pro- and eukaryotic organisms. It possesses antioxidant activity and plays pivotal roles in mammalian embryonic development and cellular functions. A very simple, prototypical selenoprotein W is SelW1 from Chlamydomonas. The U14C mutant of SelW1 was isolated and biophysically characterized. It contains an intramolecular disulfide bond and is thermally stable up to 70 °C. NMR resonance assignment of reduced and oxidized SelW1 showed that SelW1 adopts a thioredoxin fold. Interestingly, both forms show two additional sets of resonance for amino acid residues near the termini and have basically identical dynamic behavior. Since SelW1 from Chlamydomonas resembles the ancestor of mammalian selenoproteins in certain aspects, this study lays the basis for future characterization of SelW1 function and possible interaction partners.


Asunto(s)
Chlamydomonas reinhardtii/metabolismo , Mutación , Selenoproteína W/química , Selenoproteína W/metabolismo , Proteínas Algáceas/química , Proteínas Algáceas/genética , Proteínas Algáceas/metabolismo , Chlamydomonas reinhardtii/química , Chlamydomonas reinhardtii/genética , Disulfuros/química , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Oxidación-Reducción , Estabilidad Proteica , Estructura Secundaria de Proteína , Selenoproteína W/genética , Termodinámica
11.
Nat Commun ; 12(1): 2258, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859201

RESUMEN

Selenoproteins containing selenium in the form of selenocysteine are critical for bone remodeling. However, their underlying mechanism of action is not fully understood. Herein, we report the identification of selenoprotein W (SELENOW) through large-scale mRNA profiling of receptor activator of nuclear factor (NF)-κΒ ligand (RANKL)-induced osteoclast differentiation, as a protein that is downregulated via RANKL/RANK/tumour necrosis factor receptor-associated factor 6/p38 signaling. RNA-sequencing analysis revealed that SELENOW regulates osteoclastogenic genes. SELENOW overexpression enhances osteoclastogenesis in vitro via nuclear translocation of NF-κB and nuclear factor of activated T-cells cytoplasmic 1 mediated by 14-3-3γ, whereas its deficiency suppresses osteoclast formation. SELENOW-deficient and SELENOW-overexpressing mice exhibit high bone mass phenotype and osteoporosis, respectively. Ectopic SELENOW expression stimulates cell-cell fusion critical for osteoclast maturation as well as bone resorption. Thus, RANKL-dependent repression of SELENOW regulates osteoclast differentiation and blocks osteoporosis caused by overactive osteoclasts. These findings demonstrate a biological link between selenium and bone metabolism.


Asunto(s)
Remodelación Ósea/genética , Osteoclastos/fisiología , Osteogénesis/genética , Osteoporosis/genética , Selenoproteína W/metabolismo , Proteínas 14-3-3/metabolismo , Animales , Diferenciación Celular/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/fisiología , Humanos , Masculino , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/metabolismo , Osteoporosis/patología , Ligando RANK/metabolismo , RNA-Seq , Selenoproteína W/genética , Transducción de Señal/fisiología , Factor 6 Asociado a Receptor de TNF/metabolismo
12.
PLoS One ; 15(1): e0224715, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31914121

RESUMEN

Habits are automated behaviors that are insensitive to changes in behavioral outcomes. Habitual responding is thought to be mediated by the striatum, with medial striatum guiding goal-directed action and lateral striatum promoting habits. However, interspersed throughout the striatum are neurochemically differing subcompartments known as patches, which are characterized by distinct molecular profiles relative to the surrounding matrix tissue. These structures have been thoroughly characterized neurochemically and anatomically, but little is known regarding their function. Patches have been shown to be selectively activated during inflexible motor stereotypies elicited by stimulants, suggesting that patches may subserve habitual behaviors. To explore this possibility, we utilized transgenic mice (Sepw1 NP67) preferentially expressing Cre recombinase in striatal patch neurons to target these neurons for ablation with a virus driving Cre-dependent expression of caspase 3. Mice were then trained to press a lever for sucrose rewards on a variable interval schedule to elicit habitual responding. Mice were not impaired on the acquisition of this task, but lesioning striatal patches disrupted behavioral stability across training, and lesioned mice utilized a more goal-directed behavioral strategy during training. Similarly, when mice were forced to omit responses to receive sucrose rewards, habitual responding was impaired in lesioned mice. To rule out effects of lesion on motor behaviors, mice were then tested for impairments in motor learning on a rotarod and locomotion in an open field. We found that patch lesions partially impaired initial performance on the rotarod without modifying locomotor behaviors in open field. This work indicates that patches promote behavioral stability and habitual responding, adding to a growing literature implicating striatal patches in stimulus-response behaviors.


Asunto(s)
Conducta Animal/fisiología , Cuerpo Estriado/fisiología , Neostriado/fisiología , Neuronas/fisiología , Animales , Caspasa 3/genética , Estimulantes del Sistema Nervioso Central/farmacología , Dopamina/metabolismo , Hábitos , Integrasas/genética , Locomoción/fisiología , Masculino , Ratones , Ratones Transgénicos , Motivación/genética , Motivación/fisiología , Selenoproteína W
13.
Am J Physiol Cell Physiol ; 317(6): C1172-C1182, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31509445

RESUMEN

Selenoprotein W (SelW) is a selenium-containing protein with a redox motif found abundantly in the skeletal muscle of rodents. Previous in vitro studies suggest that SelW plays an antioxidant role; however, relatively few in vivo studies have addressed the antioxidant role of SelW. Since oxidative stress is a causative factor for the development of insulin resistance in obese subjects, we hypothesized that if SelW plays a role as an antioxidant, SelW deficiency could aggravate the oxidative stress and insulin resistance caused by a high-fat diet. SelW deficiency did not affect insulin sensitivity and H2O2 levels in the skeletal muscle of control diet-fed mice. SelW levels in the skeletal muscle were decreased by high-fat diet feeding for 12 wk. High-fat diet induced obesity and insulin resistance and increased the levels of H2O2 and oxidative stress makers, which were not affected by SelW deficiency. High-fat diet feeding increased the expression of antioxidant enzymes; however, SelW deficiency did not affect the expression levels of antioxidants. These results suggest that SelW does not play a protective role against oxidative stress and insulin resistance in the skeletal muscle of high-fat diet-fed obese mice.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Músculo Esquelético/metabolismo , Obesidad/genética , Estrés Oxidativo , Selenoproteína W/genética , Animales , Catalasa/genética , Catalasa/metabolismo , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Humanos , Peróxido de Hidrógeno/metabolismo , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/patología , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Selenoproteína W/deficiencia , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo
14.
Free Radic Biol Med ; 141: 362-371, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31299423

RESUMEN

Mouse selenoprotein W (SELENOW) is a small protein containing a selenocysteine (Sec, U) and four cysteine (Cys, C) residues. The Sec residue in SELENOW is located within the conserved CXXU motif corresponding to the CXXC redox motif of thioredoxin (Trx). It is known that glutathione (GSH) binds to SELENOW and that this binding is involved in protecting cells from oxidative stress. However, the regulatory mechanisms controlling the glutathionylation of SELENOW in oxidative stress are unclear. In this study, using purified recombinant SELENOW in which Sec13 was changed to Cys, we found that SELENOW was glutathionylated at Cys33 and that this S-glutathionylation was enhanced by oxidative stress. We also found that the S-glutathionylation of SELENOW at Cys33 in HEK293 cells was due to glutathione S-transferase Pi (GSTpi) and that this modification was reversed by glutaredoxin1 (Grx1). In addition to the disulfide bond between the Cys10 and Cys13 of SELENOW, a second disulfide bond was formed between Cys33 and Cys87 under oxidative stress conditions. The second disulfide bond was reduced by Trx1, but the disulfide bond between Cys10 and Cys13 was not. The second disulfide bond was also reduced by glutathione, but the disulfide bond in the CXXC motif was not. The second disulfide bond of the mutant SELENOW, in which Cys37 was replaced with Ser, was formed at a much lower concentration of hydrogen peroxide than the wild type. We also observed that Cys37 was required for S-glutathionylation, and that S-glutathionylated SELENOW containing Cys37 protected the cells from oxidative stress. Furthermore, the SELENOW (C33, 87S) mutant, which could not form the second disulfide bond, also showed antioxidant activity. Taken together, these results indicate that GSTpi-mediated S-glutathionylation of mouse SELENOW at Cys33 is required for the protection of cells in conditions of oxidative stress, through inhibition of the formation of the second disulfide bond.


Asunto(s)
Disulfuros/metabolismo , Gutatión-S-Transferasa pi/genética , Estrés Oxidativo/genética , Selenoproteína W/genética , Animales , Sitios de Unión/genética , Muerte Celular/genética , Cisteína/genética , Disulfuros/antagonistas & inhibidores , Glutarredoxinas/genética , Glutatión/genética , Glutatión/metabolismo , Células HEK293 , Humanos , Ratones , Oxidación-Reducción , Unión Proteica/genética , Selenocisteína/genética , Selenoproteína W/metabolismo
15.
Biochem Biophys Res Commun ; 512(1): 137-143, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30876693

RESUMEN

Selenoprotien W (SelW) plays a key role in brain development, although the exact biological function and mechanisms remain unclear. We performed a yeast two-hybrid screen on a human fetal brain cDNA library and identified FAM96B as a novel binding partner of SelW. FRET analyses confirmed the interaction between SelW' and FAM96B. The mutated SelW' construct was cloned and overexpressed in E. coli, and a pull-down assay verified a direct interaction between SelW' and FAM96B. Finally, Co-Immunoprecipitation on murine brain tissue proteins demonstrated an endogenous interaction between the two proteins in the brain. Taken together, our findings prove a direct interaction between SelW and FAM96B, which may provide new insights into the role of SelW in brain development and neurodegenerative diseases.


Asunto(s)
Encéfalo/metabolismo , Metaloproteínas/metabolismo , Proteínas Nucleares/metabolismo , Selenoproteína W/metabolismo , Animales , Femenino , Feto/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Biblioteca de Genes , Células HEK293 , Humanos , Metaloproteínas/genética , Ratones , Mutación , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Selenoproteína W/genética , Técnicas del Sistema de Dos Híbridos
16.
Mol Cancer ; 18(1): 9, 2019 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-30636640

RESUMEN

BACKGROUND: Breast cancer is one of the most common malignancies and the major cause of cancer-related death in women. Although the importance of PIWI-interacting RNAs (piRNAs) in cancer has been increasingly recognized, few studies have been explored the functional mechanism of piRNAs in breast cancer development and progression. METHODS: We examined the top 20 highly expressed piRNAs based on the analysis of TCGA breast cancer data in two patient cohorts to test the roles of piRNAs in breast cancer. The effects of piRNA-36,712 on the malignant phenotypes and chemosensitivity of breast cancer cells were detected in vitro and in vivo. MS2-RIP and reporter gene assays were conducted to identify the interaction and regulation among piRNA-36,712, miRNAs and SEPW1P. Kaplan-Meier estimate with log-rank test was used to compare patient survival by different piRNA-36,712 expression levels. RESULTS: We found piRNA-36,712 level was significantly lower in breast cancer than in normal breast tissues and low level was correlated with poor clinical outcome in patients. Functional studies demonstrated that piRNA-36,712 interacts with RNAs produced by SEPW1P, a retroprocessed pseudogene of SEPW1, and subsequently inhibits SEPW1 expression through competition of SEPW1 mRNA with SEPW1P RNA for microRNA-7 and microRNA-324. We also found that higher SEPW1 expression due to downregulation of piRNA-36,712 in breast cancer may suppress P53, leading to the upregulated Slug but decreased P21 and E-cadherin levels, thus promoting cancer cell proliferation, invasion and migration. Furthermore, we found that piRNA-36,712 had synergistic anticancer effects with the paclitaxel and doxorubicin, two chemotherapeutic agents for breast cancer. CONCLUSIONS: These findings suggest that piRNA-36,712 is a novel tumor suppressor and may serve as a potential predictor for the prognosis of breast cancer patients.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , ARN Interferente Pequeño/genética , Selenoproteína W/genética , Animales , Mama/efectos de los fármacos , Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Progresión de la Enfermedad , Regulación hacia Abajo , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Femenino , Humanos , MicroARNs/genética , Paclitaxel/farmacología , Pronóstico , Seudogenes , ARN Mensajero/genética , ARN Interferente Pequeño/biosíntesis , Regulación hacia Arriba
17.
Mol Biol (Mosk) ; 52(3): 519-526, 2018.
Artículo en Ruso | MEDLINE | ID: mdl-29989584

RESUMEN

Selenium is an essential trace element, the deficiency of which leads to the development of several serious diseases, including male infertility, prostate cancer, etc. It has been shown that oxidative stress contributes to the progression of prostate cancer, and antioxidants such as selenium and vitamin E can significantly reduce the risk of this disease. Sodium selenite, one of the selenium compounds that induce the formation of reactive oxygen species, is considered as a potential anticancer agent. The SS concentrations that lead to a decrease in the viability of human prostate adenocarcinoma cells (line Du-145) have been selected, and the effect of sodium selenite on the expression of mRNA of the SELV, SELW, and TGR selenocysteine proteins in these cells has been analyzed.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Neoplasias/biosíntesis , Neoplasias de la Próstata/metabolismo , Selenoproteína W/biosíntesis , Selenito de Sodio/farmacología , Línea Celular Tumoral , Humanos , Masculino , Neoplasias de la Próstata/patología
18.
Cancer Res ; 78(17): 5124-5134, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29986997

RESUMEN

Brain metastases (BM) result from the spread of primary tumors to the brain and are a leading cause of cancer mortality in adults. Secondary tissue colonization remains the main bottleneck in metastatic development, yet this "premetastatic" stage of the metastatic cascade, when primary tumor cells cross the blood-brain barrier and seed the brain before initiating a secondary tumor, remains poorly characterized. Current studies rely on specimens from fully developed macrometastases to identify therapeutic options in cancer treatment, overlooking the potentially more treatable "premetastatic" phase when colonizing cancer cells could be targeted before they initiate the secondary brain tumor. Here we use our established brain metastasis initiating cell (BMIC) models and gene expression analyses to characterize premetastasis in human lung-to-BM. Premetastatic BMIC engaged invasive and epithelial developmental mechanisms while simultaneously impeding proliferation and apoptosis. We identified the dopamine agonist apomorphine to be a potential premetastasis-targeting drug. In vivo treatment with apomorphine prevented BM formation, potentially by targeting premetastasis-associated genes KIF16B, SEPW1, and TESK2 Low expression of these genes was associated with poor survival of patients with lung adenocarcinoma. These results illuminate the cellular and molecular dynamics of premetastasis, which is subclinical and currently impossible to identify or interrogate in human patients with BM. These data present several novel therapeutic targets and associated pathways to prevent BM initiation.Significance: These findings unveil molecular features of the premetastatic stage of lung-to-brain metastases and offer a potential therapeutic strategy to prevent brain metastases. Cancer Res; 78(17); 5124-34. ©2018 AACR.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Terapia Molecular Dirigida , Metástasis de la Neoplasia/tratamiento farmacológico , Apomorfina/farmacología , Apoptosis/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Dopamina/metabolismo , Agonistas de Dopamina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Cinesinas/genética , Pulmón/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Proteínas Serina-Treonina Quinasas/genética , Selenoproteína W/genética
19.
ACS Chem Neurosci ; 9(7): 1560-1565, 2018 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-29714059

RESUMEN

Abnormal accumulation of tau protein into oligomers contributes to neuronal dysfunction. Reduction of tau level is potentially able to prevent its accumulation. Here we uncover a critical role of the free thiol at Cys-322 in determining tau stability. We found that the application of thiol-blocking agents like NEM or MMTS blocks this thiol, by which it destabilizes tau protein and prevents its oligomer formation. Furthermore, we identified a tau-interacting protein, selenoprotein W, which attenuates tau accumulation by forming disulfide linkage between SelW Cys-37 and tau Cys-322. These findings provide a promising strategy to prevent tau accumulation and oligomer formation.


Asunto(s)
Proteínas tau/química , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Apoptosis , Encéfalo/metabolismo , Cisteína/metabolismo , Escherichia coli , Células HEK293 , Humanos , Peróxido de Hidrógeno , Ratones Transgénicos , Agregación Patológica de Proteínas/tratamiento farmacológico , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Estabilidad Proteica/efectos de los fármacos , Proteínas Recombinantes/química , Selenoproteína W/metabolismo , Compuestos de Sulfhidrilo/antagonistas & inhibidores , Compuestos de Sulfhidrilo/metabolismo , Proteínas tau/genética
20.
Metallomics ; 10(5): 751-758, 2018 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-29766197

RESUMEN

Selenoprotein W (SelW) is an important member of the avian selenoprotein family. It is well known for its important role in protecting neurons from oxidative stress during neuronal development. d-Amino acid (d-serine), as a neurotransmitter in the central nervous system (CNS), can mediate neurotoxicity. d-Amino acid oxidase (DAAO) is responsible for regulating the d-serine levels in cells. However, the correlation between SelW and DAAO is not clear yet. To investigate the regulations between SelW and DAAO, chicken embryo monolayer neurons were treated with d-serine and/or Se. In this study, we predicted molecular binding between SelW and DAAO. These results showed that the 9-16, 18, 41-47 and 66 residues of SelW could combine with the DAAO, which suggested that chicken SelW might be the target of DAAO. We determined the DAAO activity and the mRNA expression of SelW in in vitro cultured chicken embryo primitive neuron cells. d-Serine influenced the activity of DAAO and, moreover, a significant increase in the mRNA expression of SelW was found in neurons treated with Se. Notably, we also observed changes in the expression of SelW and DAAO when neurons were treated with various concentrations of d-serine and Se. In conclusion, these data suggest that d-serine could regulate the mRNA expression of SelW by interfering with the activity of DAAO in chicken embryo neurons.


Asunto(s)
D-Aminoácido Oxidasa/metabolismo , Regulación de la Expresión Génica , Neuronas/metabolismo , Selenoproteína W/metabolismo , Serina/farmacología , Animales , Supervivencia Celular , Células Cultivadas , Embrión de Pollo , Pollos , D-Aminoácido Oxidasa/genética , Proyección Neuronal , Neuronas/citología , Neuronas/efectos de los fármacos , Estrés Oxidativo , Selenio/farmacología , Selenoproteína W/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...